Mol. Cells 2022; 45(4): 202-215
Published online January 3, 2022
https://doi.org/10.14348/molcells.2021.0206
© The Korean Society for Molecular and Cellular Biology
Correspondence to : echo@skku.edu
This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/.
The androgen receptor (AR) is an important therapeutic target for treating prostate cancer (PCa). Moreover, there is an increasing need for understanding the AR-independent progression of tumor cells such as neuroendocrine prostate cancer (NEPC). Menin, which is encoded by multiple endocrine neoplasia type 1 (MEN1), serves as a direct link between AR and the mixed-lineage leukemia (MLL) complex in PCa development by activating AR target genes through histone H3 lysine 4 methylation. Although menin is a critical component of AR signaling, its tumorigenic role in AR-independent PCa cells remains unknown. Here, we compared the role of menin in AR-positive and AR-negative PCa cells via RNAi-mediated or pharmacological inhibition of menin. We demonstrated that menin was involved in tumor cell growth and metastasis in PCa cells with low or deficient levels of AR. The inhibition of menin significantly diminished the growth of PCa cells and induced apoptosis, regardless of the presence of AR. Additionally, transcriptome analysis showed that the expression of many metastasis-associated genes was perturbed by menin inhibition in AR-negative DU145 cells. Furthermore, wound-healing assay results showed that menin promoted cell migration in AR-independent cellular contexts. Overall, these findings suggest a critical function of menin in tumorigenesis and provide a rationale for drug development against menin toward targeting high-risk metastatic PCa, especially those independent of AR.
Keywords androgen receptor, menin, menin inhibitor, metastasis, prostate cancer
Alternatively, menin displays an oncogenic function in other cell types, such as blood cells. Mixed-lineage leukemia (MLL) is an extremely aggressive blood cancer characterized by MLL fusion oncogenes generated by the chromosomal translocation with genes involved in the transcription elongation (Slany, 2009). The most prominent binding partner of menin is MLL, which has a SET domain-derived methyltransferase activity toward histone H3 lysine 4 (H3K4). As an integral part of the MLL1/MLL2 complex, menin tethers MLL complexes to promote H3K4 trimethylation (H3K4me3) and gene activation (Hughes et al., 2004; Rao and Dou, 2015). In MLL-rearranged leukemia, menin recruits MLL fusion protein to target genes together with lens epithelium-derived growth factor. Menin-mediated chromatin remodeling via H3K4me3 and Dot1L-dependent H3K79me3 increases the transcription of
In solid tumors, aberrantly expressed
Furthermore, menin and MLL participate in the androgen receptor (AR) pathway and play an essential role in the development of castration-resistance prostate cancer (CRPC) (Malik et al., 2015). CRPC develops by reactivated AR signaling via
However, the many incidences of AR-independent PCa growth indicate alternative AR-bypass mechanisms during disease progression. For example, neuroendocrine prostate cancer (NEPC) is highly aggressive and characterized by resistance to AR-targeted therapy due to its independence from AR signaling. Additionally, emerging lines of evidence suggest that menin plays diverse roles in tumorigenesis independently of MLL (Chou et al., 2020; Feng et al., 2017b; Gang et al., 2013). For instance, menin directly interacts with Myc in fibrosarcoma cells to upregulate Myc target genes in an MLL-independent manner (Wu et al., 2017).
In this study, we elucidated the role of menin in metastasis and proliferation in AR-independent tumorigenesis. The results of our study expand the potential therapeutic scope of existing limited therapeutic options, especially for AR-negative PCa.
Human PCa DU145 cells from the American Type Culture Collection (ATCC, USA) were maintained in MEM media (Welgene, Korea) supplemented with 10% fetal bovine serum (FBS) (Thermo Fisher Scientific, USA), 100 U/ml penicillin, and 100 μg/ml streptomycin (Welgene) at 37°C with 5% CO2. LNCaP (ATCC) and 22RV1 (ATCC) cells were maintained in RPMI-1640 media (Welgene) supplemented with 10% FBS (Thermo Fisher Scientific), 100 U/ml penicillin, and 100 μg/ml streptomycin (Welgene) at 37°C with 5% CO2. PC3 (ATCC) cells were maintained in F12-K media (Welgene) supplemented with 10% FBS (Thermo Fisher Scientific), 100 U/ml penicillin, and 100 μg/ml streptomycin (Welgene) at 37°C with 5% CO2. LNCaP and 22RV1 cells are AR-positive, whereas DU145 and PC3 cells are AR-negative.
A cell suspension aliquot was centrifuged at 800 rpm for 3 min, resuspended in 1 ml phosphate-buffered saline (PBS), and mixed with 0.4% trypan blue (Thermo Fisher Scientific) prior to loading a hemocytometer. The number of unstained (viable) and stained (nonviable) cells were counted using a binocular microscope. The percentage of unstained cells was calculated as follows: viable cells (%) = (number of viable cells per ml / number of cells per ml) × 100.
DU145 and LNCaP cells were seeded at 2,000 cells/well and 5,000 cells/well, respectively, in 96-well plates and treated with different concentrations of MI-503 for 96 h. Cell proliferation was determined using Cell Counting Kit-8 (CCK-8) from Dojindo Laboratories (USA) and CellTiter-Glo from Promega (USA). Twenty microliters of CCK-8 reagent were added to each well, and plates were incubated for 1-4 h at 37°C. Absorbance was measured at 490 nm using a multimode Synergy HTX reader (BioTek, USA). To detect viable cells, an ATP assay was performed with CellTiter-Glo. First, 100 μl of CellTiter-Glo reagent was added to each well, followed by incubation at room temperature for 10 min. The luminescence signal was recorded, and the half-maximal inhibitory concentration (IC50) was determined from the derived dose-response curve.
Cells were transfected with non-targeting control or specific siRNAs at concentrations ranging from 60 to 100 nM using Lipofectamine RNAiMAX (Thermo Fisher Scientific), according to the manufacturer’s instructions. After transfection, cells were incubated for 72 h. The efficiency of siRNA-mediated knockdown was determined by quantitative real-time polymerase chain reaction (RT-qPCR) and Western blotting. siRNA sequences are shown in Supplementary Table S1.
Total RNA was extracted using a NucleoSpin® RNA kit (Macherey-Nagel, USA), according to the manufacturer’s instructions. Approximately 2 μg of extracted RNA was used for cDNA synthesis with a cDNA synthesis kit (Thermo Fisher Scientific). RT-qPCR was performed using KAPA SYBR FAST Master Mix (KAPA Biosystem, USA) and a CFX96 Real-Time System (Bio-Rad Laboratories, USA) with specific primers (Supplementary Table S2). The relative quantification was calculated using the 2-ΔΔCT (Livak) method (Livak and Schmittgen, 2001) and the relative levels of mRNA were normalized to those of β-actin for each reaction.
Cells were harvested, washed twice with cold PBS, and lysed using a lysis buffer consisting of 25 mM Tris HCl (pH 8.0), 1 mM EDTA, 150 mM NaCl, and 0.5% NP40. After a 10-min incubation on ice, the cell lysate was centrifuged at 13,000 rpm at 4°C for 10 min. Equal amounts of protein from extracts were separated using SDS-PAGE and transferred to nitrocellulose membranes. The blots were blocked with 5% skim milk and incubated with primary antibodies, which were diluted in the range of 1:1,000 to 1:5,000. After washing with TBS-T (5 mM Tris HCl [pH 7.4], 150 mM NaCl, and 0.01% Tween 20), secondary antibodies were added at a dilution of 1:10,000 for a 1-h incubation at room temperature. Protein signals were detected using a western blot detection kit (AbFrontier, Korea). The following primary antibodies were used in this study: anti-β-actin (Merck Millipore, USA), anti-α-tubulin (AbFrontier), anti-menin (Bethyl Laboratories, USA), anti-cleaved-PARP (Cell Signaling Technology, USA), anti-AR (Santa Cruz Biotechnology, USA), and anti-Lamin B1 (Thermo Fisher Scientific).
Cells grown in 6-well plates were treated with appropriate siRNAs or MI-503 for 48 h. Pelleted cells were washed thoroughly with PBS and resuspended in annexin V-FITC and propidium iodide (PI) at 50 μg/ml in binding buffer. After incubation at room temperature in the dark for 15 min, cells were detected using a Guava® easyCyte Flow Cytometer (Merck Millipore).
Cells were fixed with 4% (w/v) paraformaldehyde, permeabilized with 0.5% Triton X-100 (Sigma, USA) in PBS, and blocked with 2% (w/v) bovine serum albumin in PBS. Cell nuclei were stained with DAPI (Thermo Fisher Scientific). Cells were examined under a LSM700 confocal microscope (Carl Zeiss, Germany), and images were processed using ZEISS ZEN Software.
Total RNA was extracted using TRIzol® (Thermo Fisher Scientific) and checked for integrity using a RNA 6000 Nano ll kit and Bioanalyzer 2100 (Agilent, USA). A RNA library was prepared using a NEXTflex® Rapid Directional mRNA-Seq kit (Bio Scientific, USA). Briefly, mRNA was isolated from 400 ng of total RNA using RNA purification beads by polyA capture, followed by enzyme shearing. After cDNA synthesis, A-tailing and end repair were performed to ligate the cDNA to primers containing unique sequencing adaptors with an index for tracking Illumina reads. For each library, the total library size (~400 bp) was confirmed using a Bioanalyzer and High Sensitivity DNA Chips (Agilent) and quantified by RT-qPCR using a CFX96 Real-Time System (Bio-Rad Laboratories). After normalization, sequencing was performed using an Illumina NextSeq 500. Clusters of cDNA libraries were generated on a flow cell and sequenced generating 75-bp paired-end reads (2 × 75) using a NextSeq 500 and 150-Cycle Reagent Cartridge kit (Illumina, USA). Raw images were transformed by base-calling into sequence data and were stored in the FASTQ format.
Cells were seeded in 6-well plates, incubated for 24 h, transfected with control or specific siRNAs using Lipofectamine 2000 (Thermo Fisher Scientific), and incubated for another 48 h until confluent. Cells were then scratched with a pin (SPL, Korea), and the detached cells were removed to create a cell-free zone as a thin wound. Cell migration was observed immediately and 24 h after wounding using a microscope.
DU145 cells were cross-linked with 1% formaldehyde at room temperature for 12.5 min, followed by quenching with 0.125 M glycine. Nuclei were prepared with Buffer A, containing 10 mM Tris HCl (pH 7.5), 10 mM KCl, 5 mM MgCl2, and 0.5% NP40, and resuspended in Buffer B, consisting of 50 mM Tris HCl (pH 7.9), 10 mM EDTA, and 0.5% SDS. The chromatin solution was prepared by sonication using a Bioruptor (Cosmo Bio, Japan). The supernatant of the sonicated nuclei was diluted tenfold in ChIP dilution buffer and used for immunoprecipitation with indicated antibodies. ChIP was performed by incubating the chromatin solution with 7.5 μg of anti-menin antibody (Bethyl Laboratories), 4 μg of H3K9 me3 antibody (Active Motif, USA), 2 μg of H3 antibody (Active Motif), or 2 μg of rabbit IgG (Thermo Fisher Scientific) as a control, followed by recovery using Protein A Sepharose beads and a 2-h incubation. Beads were washed sequentially with a low-salt wash buffer, high-salt wash buffer, LiCl wash buffer, and finally TE buffer (50 mM Tris HCl and 10 mM EDTA) twice. The precipitated DNA was resuspended in ChIP elution buffer. Cross-linking was reversed for 4 h at 65°C in the presence of RNase A. The precipitated DNA was then purified using a PCR purification kit (Qiagen, Germany) and measured using qPCR and specific ChIP-qPCR primers (Supplementary Table S3). The calculated input (%) was determined as follows: (immunoprecipitated DNA / input DNA) × 100, and represented the relative enrichment of immunoprecipitated DNA.
The publicly available mRNA expression datasets from healthy controls and patients with corresponding cancers in GEO (https://www.ncbi.nlm.nih.gov/geo/) were analyzed. The gene expression levels of 497 patients with PCa from TCGA were log2 transformed and then analyzed. TCGA disease-free survival data and normalized mRNA expression levels (RSEM, batch normalized from Illumina HiSeq_RNASeqV2) were downloaded from the cBioPortal for Cancer Genomics (http://www.cbioportal.org). Kaplan–Meier analysis and log-rank tests were performed using the survival package in R.
Data were reported as mean from three biological replicates unless otherwise stated in figure legends. Error bars represent the SD. The Student’s
Menin is a critical component of AR signaling, but its tumorigenic role in AR-negative PCa cells remains unknown. To elucidate the activity of menin in the AR-independent pathway, we first investigated whether the inhibition of menin affects the growth of AR-negative and AR-positive PCa cells. In this regard, the DU145, PC3, LNCaP, and 22RV1 cell lines were used as a preclinical model of PCa for comparison. DU145 and PC3 are AR-negative and androgen unresponsive PCa cells, whereas LNCaP and 22RV1 are AR-positive and androgen responsive PCa cells. These cell lines showed comparable levels of menin expression (Fig. 1A). Cells were treated with control or two independent
The inhibition of menin increases apoptosis in Rb-deficient thyroid carcinoma and MLL-rearranged leukemia cells (Grembecka et al., 2012; Matoso et al., 2008; Shin et al., 2016). Considering the inhibitory effect of menin depletion on cell growth, we also compared the effect of menin inhibition on apoptotic cell death. FACS analysis with annexin V-FITC/PI double staining showed a significant increase in apoptotic signals, indicating that cell apoptosis was induced by the transfection of
To assess the effect of menin depletion on gene expression, we performed gene expression analysis with DU145 and LNCaP cells transfected with
We further dissected the role of menin in AR-negative cells by analyzing the 880 DU145-specific DEGs. Surprisingly, our GO analysis showed that the genes perturbed in DU145 cells, are significantly associated with biological functions indicative of metastasis, such as cell adhesion, angiogenesis, negative regulation of cell proliferation, and extracellular matrix disassembly/organization (Fig. 3D). Representative genes upregulated by menin depletion are listed in Fig. 3E. Together, these data demonstrate that menin plays an unexpected role in enhancing the metastatic potential in low or null AR PCa cells.
To further explore the potential role of menin in metastatic PCa, we evaluated the expression profiles of
To test a potential link between menin and metastasis, we next investigated whether menin affects tumor cell motility in AR-negative cells. We performed a scratch wound-healing assay to evaluate the migration capability of DU145 and PC3, following transfection with control or
Cell migration is an essential process involved in cancer metastasis and is driven by a complex transcriptional program. To explore the AR-independent function of menin in metastasis, we further focused on DU145-specific DEGs that showed connectivity to metastasis. Some metastasis-related genes, including
To gain further insights into the function of menin in metastatic pathways, we focused on
To explore the molecular mechanisms of menin-dependent
Next, we explored any potential link between menin and SUV39H1 in the regulation of metastasis-related genes. We determined whether the expression of
NEPC is a subtype of PCa with poor prognosis. NEPC can arise from resistance to AR-targeted hormonal therapy or
Menin is strongly associated with the tumorigenesis of various cancers, functioning as a direct coactivator of many prognostic transcriptional factors through the tethering of MLL complexes to chromatin. Thus, the menin-MLL interface is a valuable target for anticancer therapy (Grembecka et al., 2012; Huang et al., 2012; Xu et al., 2016). Menin and MLL are also involved in AR signaling as coactivators to promote transcriptional activation of AR target genes in prostate tumors. In this study, using TCGA and GEO analysis, we showed that the level of menin was significantly elevated in PCa and even higher in metastatic cancers. These findings suggest that a high level of menin is related to metastasis and the negative prognosis of patients with PCa. Furthermore, we observed that treating AR-negative cells with
In particular, the analysis of DU145-specific DEGs revealed the downstream target genes of menin as associated with metastasis. MMPs and tissue inhibitors of metalloproteinases (TIMPs) are well-known players of tissue homeostasis and controllers of tumor behavior in various cancer types (Reis et al., 2012; Ross et al., 2003). MMPs are responsible for the degradation of the ECM to facilitate malignant tumor invasion and metastasis. For example, MMP2 cleaves the basal components of ECM, such as type IV collagen and gelatin. MMPs are mainly regulated at the posttranslational level by TIMPs, which prevent the substrate interaction of MMPs by binding proximally to their catalytic domain. Therefore, the decreased expression of TIMPs may enhance MMP activity, thereby promoting tumor growth and invasiveness.
Importantly, we showed that menin is directly associated with the
The AR signaling axis is a critical part of tumor growth and metastatic development of PCa (Ko et al., 2015; Lin et al., 2018). However, unexpectedly, our migration assay showed that menin depletion affected the migratory potential of AR-negative DU145 and PC3 cells. At the molecular level, menin was linked to the regulation of metastasis-associated genes in DU145 cells, indicating that menin promotes metastatic behavior of tumor cells through the regulation of metastasis genes in an AR-independent manner. However, the underlying mechanisms and affected genes are expected to be distinct for each cell line. For example, genes like
In summary, our study shows an unexpected role of menin in the metastasis of PCa that does not depend on AR signaling. In light of our findings, menin may be a future target for small molecule inhibitors and act as a prognostic biomarker for PCa metastasis.
This work was supported by National Research Foundation of Korea (NRF) grants funded by the Korean government (NRF-2019R1A5A2027340, 2021R1F1A1049941, and 2019R1A2C2084716 to E.J.C. and 2012R1A3A2048767 to H.D.Y.).
T.K., K.J., E.K., K.Y., J.H.P., and J.C. performed the experiments, analyzed the data, wrote the manuscript. J.C. and J.H.K. performed bioinformatics. H.D.Y. and H.S.K. provided reagents. E.J.C. conceived the study design and wrote the manuscript. T.K. and K.J. contributed equally to this work.
The authors have no potential conflicts of interest to disclose.
Mol. Cells 2022; 45(4): 202-215
Published online April 30, 2022 https://doi.org/10.14348/molcells.2021.0206
Copyright © The Korean Society for Molecular and Cellular Biology.
Taewan Kim1,4 , Kwanyoung Jeong1,4
, Eunji Kim1
, Kwanghyun Yoon1
, Jinmi Choi1
, Jae Hyeon Park1
, Jae-Hwan Kim2,3
, Hyung Sik Kim1
, Hong-Duk Youn3
, and Eun-Jung Cho1,*
1School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea, 2NineBiopharm, Co., Ltd., Cheongju 28161, Korea, 3National Creative Research Center for Epigenome Reprogramming Network, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea, 4These authors contributed equally to this work.
Correspondence to:echo@skku.edu
This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/.
The androgen receptor (AR) is an important therapeutic target for treating prostate cancer (PCa). Moreover, there is an increasing need for understanding the AR-independent progression of tumor cells such as neuroendocrine prostate cancer (NEPC). Menin, which is encoded by multiple endocrine neoplasia type 1 (MEN1), serves as a direct link between AR and the mixed-lineage leukemia (MLL) complex in PCa development by activating AR target genes through histone H3 lysine 4 methylation. Although menin is a critical component of AR signaling, its tumorigenic role in AR-independent PCa cells remains unknown. Here, we compared the role of menin in AR-positive and AR-negative PCa cells via RNAi-mediated or pharmacological inhibition of menin. We demonstrated that menin was involved in tumor cell growth and metastasis in PCa cells with low or deficient levels of AR. The inhibition of menin significantly diminished the growth of PCa cells and induced apoptosis, regardless of the presence of AR. Additionally, transcriptome analysis showed that the expression of many metastasis-associated genes was perturbed by menin inhibition in AR-negative DU145 cells. Furthermore, wound-healing assay results showed that menin promoted cell migration in AR-independent cellular contexts. Overall, these findings suggest a critical function of menin in tumorigenesis and provide a rationale for drug development against menin toward targeting high-risk metastatic PCa, especially those independent of AR.
Keywords: androgen receptor, menin, menin inhibitor, metastasis, prostate cancer
Alternatively, menin displays an oncogenic function in other cell types, such as blood cells. Mixed-lineage leukemia (MLL) is an extremely aggressive blood cancer characterized by MLL fusion oncogenes generated by the chromosomal translocation with genes involved in the transcription elongation (Slany, 2009). The most prominent binding partner of menin is MLL, which has a SET domain-derived methyltransferase activity toward histone H3 lysine 4 (H3K4). As an integral part of the MLL1/MLL2 complex, menin tethers MLL complexes to promote H3K4 trimethylation (H3K4me3) and gene activation (Hughes et al., 2004; Rao and Dou, 2015). In MLL-rearranged leukemia, menin recruits MLL fusion protein to target genes together with lens epithelium-derived growth factor. Menin-mediated chromatin remodeling via H3K4me3 and Dot1L-dependent H3K79me3 increases the transcription of
In solid tumors, aberrantly expressed
Furthermore, menin and MLL participate in the androgen receptor (AR) pathway and play an essential role in the development of castration-resistance prostate cancer (CRPC) (Malik et al., 2015). CRPC develops by reactivated AR signaling via
However, the many incidences of AR-independent PCa growth indicate alternative AR-bypass mechanisms during disease progression. For example, neuroendocrine prostate cancer (NEPC) is highly aggressive and characterized by resistance to AR-targeted therapy due to its independence from AR signaling. Additionally, emerging lines of evidence suggest that menin plays diverse roles in tumorigenesis independently of MLL (Chou et al., 2020; Feng et al., 2017b; Gang et al., 2013). For instance, menin directly interacts with Myc in fibrosarcoma cells to upregulate Myc target genes in an MLL-independent manner (Wu et al., 2017).
In this study, we elucidated the role of menin in metastasis and proliferation in AR-independent tumorigenesis. The results of our study expand the potential therapeutic scope of existing limited therapeutic options, especially for AR-negative PCa.
Human PCa DU145 cells from the American Type Culture Collection (ATCC, USA) were maintained in MEM media (Welgene, Korea) supplemented with 10% fetal bovine serum (FBS) (Thermo Fisher Scientific, USA), 100 U/ml penicillin, and 100 μg/ml streptomycin (Welgene) at 37°C with 5% CO2. LNCaP (ATCC) and 22RV1 (ATCC) cells were maintained in RPMI-1640 media (Welgene) supplemented with 10% FBS (Thermo Fisher Scientific), 100 U/ml penicillin, and 100 μg/ml streptomycin (Welgene) at 37°C with 5% CO2. PC3 (ATCC) cells were maintained in F12-K media (Welgene) supplemented with 10% FBS (Thermo Fisher Scientific), 100 U/ml penicillin, and 100 μg/ml streptomycin (Welgene) at 37°C with 5% CO2. LNCaP and 22RV1 cells are AR-positive, whereas DU145 and PC3 cells are AR-negative.
A cell suspension aliquot was centrifuged at 800 rpm for 3 min, resuspended in 1 ml phosphate-buffered saline (PBS), and mixed with 0.4% trypan blue (Thermo Fisher Scientific) prior to loading a hemocytometer. The number of unstained (viable) and stained (nonviable) cells were counted using a binocular microscope. The percentage of unstained cells was calculated as follows: viable cells (%) = (number of viable cells per ml / number of cells per ml) × 100.
DU145 and LNCaP cells were seeded at 2,000 cells/well and 5,000 cells/well, respectively, in 96-well plates and treated with different concentrations of MI-503 for 96 h. Cell proliferation was determined using Cell Counting Kit-8 (CCK-8) from Dojindo Laboratories (USA) and CellTiter-Glo from Promega (USA). Twenty microliters of CCK-8 reagent were added to each well, and plates were incubated for 1-4 h at 37°C. Absorbance was measured at 490 nm using a multimode Synergy HTX reader (BioTek, USA). To detect viable cells, an ATP assay was performed with CellTiter-Glo. First, 100 μl of CellTiter-Glo reagent was added to each well, followed by incubation at room temperature for 10 min. The luminescence signal was recorded, and the half-maximal inhibitory concentration (IC50) was determined from the derived dose-response curve.
Cells were transfected with non-targeting control or specific siRNAs at concentrations ranging from 60 to 100 nM using Lipofectamine RNAiMAX (Thermo Fisher Scientific), according to the manufacturer’s instructions. After transfection, cells were incubated for 72 h. The efficiency of siRNA-mediated knockdown was determined by quantitative real-time polymerase chain reaction (RT-qPCR) and Western blotting. siRNA sequences are shown in Supplementary Table S1.
Total RNA was extracted using a NucleoSpin® RNA kit (Macherey-Nagel, USA), according to the manufacturer’s instructions. Approximately 2 μg of extracted RNA was used for cDNA synthesis with a cDNA synthesis kit (Thermo Fisher Scientific). RT-qPCR was performed using KAPA SYBR FAST Master Mix (KAPA Biosystem, USA) and a CFX96 Real-Time System (Bio-Rad Laboratories, USA) with specific primers (Supplementary Table S2). The relative quantification was calculated using the 2-ΔΔCT (Livak) method (Livak and Schmittgen, 2001) and the relative levels of mRNA were normalized to those of β-actin for each reaction.
Cells were harvested, washed twice with cold PBS, and lysed using a lysis buffer consisting of 25 mM Tris HCl (pH 8.0), 1 mM EDTA, 150 mM NaCl, and 0.5% NP40. After a 10-min incubation on ice, the cell lysate was centrifuged at 13,000 rpm at 4°C for 10 min. Equal amounts of protein from extracts were separated using SDS-PAGE and transferred to nitrocellulose membranes. The blots were blocked with 5% skim milk and incubated with primary antibodies, which were diluted in the range of 1:1,000 to 1:5,000. After washing with TBS-T (5 mM Tris HCl [pH 7.4], 150 mM NaCl, and 0.01% Tween 20), secondary antibodies were added at a dilution of 1:10,000 for a 1-h incubation at room temperature. Protein signals were detected using a western blot detection kit (AbFrontier, Korea). The following primary antibodies were used in this study: anti-β-actin (Merck Millipore, USA), anti-α-tubulin (AbFrontier), anti-menin (Bethyl Laboratories, USA), anti-cleaved-PARP (Cell Signaling Technology, USA), anti-AR (Santa Cruz Biotechnology, USA), and anti-Lamin B1 (Thermo Fisher Scientific).
Cells grown in 6-well plates were treated with appropriate siRNAs or MI-503 for 48 h. Pelleted cells were washed thoroughly with PBS and resuspended in annexin V-FITC and propidium iodide (PI) at 50 μg/ml in binding buffer. After incubation at room temperature in the dark for 15 min, cells were detected using a Guava® easyCyte Flow Cytometer (Merck Millipore).
Cells were fixed with 4% (w/v) paraformaldehyde, permeabilized with 0.5% Triton X-100 (Sigma, USA) in PBS, and blocked with 2% (w/v) bovine serum albumin in PBS. Cell nuclei were stained with DAPI (Thermo Fisher Scientific). Cells were examined under a LSM700 confocal microscope (Carl Zeiss, Germany), and images were processed using ZEISS ZEN Software.
Total RNA was extracted using TRIzol® (Thermo Fisher Scientific) and checked for integrity using a RNA 6000 Nano ll kit and Bioanalyzer 2100 (Agilent, USA). A RNA library was prepared using a NEXTflex® Rapid Directional mRNA-Seq kit (Bio Scientific, USA). Briefly, mRNA was isolated from 400 ng of total RNA using RNA purification beads by polyA capture, followed by enzyme shearing. After cDNA synthesis, A-tailing and end repair were performed to ligate the cDNA to primers containing unique sequencing adaptors with an index for tracking Illumina reads. For each library, the total library size (~400 bp) was confirmed using a Bioanalyzer and High Sensitivity DNA Chips (Agilent) and quantified by RT-qPCR using a CFX96 Real-Time System (Bio-Rad Laboratories). After normalization, sequencing was performed using an Illumina NextSeq 500. Clusters of cDNA libraries were generated on a flow cell and sequenced generating 75-bp paired-end reads (2 × 75) using a NextSeq 500 and 150-Cycle Reagent Cartridge kit (Illumina, USA). Raw images were transformed by base-calling into sequence data and were stored in the FASTQ format.
Cells were seeded in 6-well plates, incubated for 24 h, transfected with control or specific siRNAs using Lipofectamine 2000 (Thermo Fisher Scientific), and incubated for another 48 h until confluent. Cells were then scratched with a pin (SPL, Korea), and the detached cells were removed to create a cell-free zone as a thin wound. Cell migration was observed immediately and 24 h after wounding using a microscope.
DU145 cells were cross-linked with 1% formaldehyde at room temperature for 12.5 min, followed by quenching with 0.125 M glycine. Nuclei were prepared with Buffer A, containing 10 mM Tris HCl (pH 7.5), 10 mM KCl, 5 mM MgCl2, and 0.5% NP40, and resuspended in Buffer B, consisting of 50 mM Tris HCl (pH 7.9), 10 mM EDTA, and 0.5% SDS. The chromatin solution was prepared by sonication using a Bioruptor (Cosmo Bio, Japan). The supernatant of the sonicated nuclei was diluted tenfold in ChIP dilution buffer and used for immunoprecipitation with indicated antibodies. ChIP was performed by incubating the chromatin solution with 7.5 μg of anti-menin antibody (Bethyl Laboratories), 4 μg of H3K9 me3 antibody (Active Motif, USA), 2 μg of H3 antibody (Active Motif), or 2 μg of rabbit IgG (Thermo Fisher Scientific) as a control, followed by recovery using Protein A Sepharose beads and a 2-h incubation. Beads were washed sequentially with a low-salt wash buffer, high-salt wash buffer, LiCl wash buffer, and finally TE buffer (50 mM Tris HCl and 10 mM EDTA) twice. The precipitated DNA was resuspended in ChIP elution buffer. Cross-linking was reversed for 4 h at 65°C in the presence of RNase A. The precipitated DNA was then purified using a PCR purification kit (Qiagen, Germany) and measured using qPCR and specific ChIP-qPCR primers (Supplementary Table S3). The calculated input (%) was determined as follows: (immunoprecipitated DNA / input DNA) × 100, and represented the relative enrichment of immunoprecipitated DNA.
The publicly available mRNA expression datasets from healthy controls and patients with corresponding cancers in GEO (https://www.ncbi.nlm.nih.gov/geo/) were analyzed. The gene expression levels of 497 patients with PCa from TCGA were log2 transformed and then analyzed. TCGA disease-free survival data and normalized mRNA expression levels (RSEM, batch normalized from Illumina HiSeq_RNASeqV2) were downloaded from the cBioPortal for Cancer Genomics (http://www.cbioportal.org). Kaplan–Meier analysis and log-rank tests were performed using the survival package in R.
Data were reported as mean from three biological replicates unless otherwise stated in figure legends. Error bars represent the SD. The Student’s
Menin is a critical component of AR signaling, but its tumorigenic role in AR-negative PCa cells remains unknown. To elucidate the activity of menin in the AR-independent pathway, we first investigated whether the inhibition of menin affects the growth of AR-negative and AR-positive PCa cells. In this regard, the DU145, PC3, LNCaP, and 22RV1 cell lines were used as a preclinical model of PCa for comparison. DU145 and PC3 are AR-negative and androgen unresponsive PCa cells, whereas LNCaP and 22RV1 are AR-positive and androgen responsive PCa cells. These cell lines showed comparable levels of menin expression (Fig. 1A). Cells were treated with control or two independent
The inhibition of menin increases apoptosis in Rb-deficient thyroid carcinoma and MLL-rearranged leukemia cells (Grembecka et al., 2012; Matoso et al., 2008; Shin et al., 2016). Considering the inhibitory effect of menin depletion on cell growth, we also compared the effect of menin inhibition on apoptotic cell death. FACS analysis with annexin V-FITC/PI double staining showed a significant increase in apoptotic signals, indicating that cell apoptosis was induced by the transfection of
To assess the effect of menin depletion on gene expression, we performed gene expression analysis with DU145 and LNCaP cells transfected with
We further dissected the role of menin in AR-negative cells by analyzing the 880 DU145-specific DEGs. Surprisingly, our GO analysis showed that the genes perturbed in DU145 cells, are significantly associated with biological functions indicative of metastasis, such as cell adhesion, angiogenesis, negative regulation of cell proliferation, and extracellular matrix disassembly/organization (Fig. 3D). Representative genes upregulated by menin depletion are listed in Fig. 3E. Together, these data demonstrate that menin plays an unexpected role in enhancing the metastatic potential in low or null AR PCa cells.
To further explore the potential role of menin in metastatic PCa, we evaluated the expression profiles of
To test a potential link between menin and metastasis, we next investigated whether menin affects tumor cell motility in AR-negative cells. We performed a scratch wound-healing assay to evaluate the migration capability of DU145 and PC3, following transfection with control or
Cell migration is an essential process involved in cancer metastasis and is driven by a complex transcriptional program. To explore the AR-independent function of menin in metastasis, we further focused on DU145-specific DEGs that showed connectivity to metastasis. Some metastasis-related genes, including
To gain further insights into the function of menin in metastatic pathways, we focused on
To explore the molecular mechanisms of menin-dependent
Next, we explored any potential link between menin and SUV39H1 in the regulation of metastasis-related genes. We determined whether the expression of
NEPC is a subtype of PCa with poor prognosis. NEPC can arise from resistance to AR-targeted hormonal therapy or
Menin is strongly associated with the tumorigenesis of various cancers, functioning as a direct coactivator of many prognostic transcriptional factors through the tethering of MLL complexes to chromatin. Thus, the menin-MLL interface is a valuable target for anticancer therapy (Grembecka et al., 2012; Huang et al., 2012; Xu et al., 2016). Menin and MLL are also involved in AR signaling as coactivators to promote transcriptional activation of AR target genes in prostate tumors. In this study, using TCGA and GEO analysis, we showed that the level of menin was significantly elevated in PCa and even higher in metastatic cancers. These findings suggest that a high level of menin is related to metastasis and the negative prognosis of patients with PCa. Furthermore, we observed that treating AR-negative cells with
In particular, the analysis of DU145-specific DEGs revealed the downstream target genes of menin as associated with metastasis. MMPs and tissue inhibitors of metalloproteinases (TIMPs) are well-known players of tissue homeostasis and controllers of tumor behavior in various cancer types (Reis et al., 2012; Ross et al., 2003). MMPs are responsible for the degradation of the ECM to facilitate malignant tumor invasion and metastasis. For example, MMP2 cleaves the basal components of ECM, such as type IV collagen and gelatin. MMPs are mainly regulated at the posttranslational level by TIMPs, which prevent the substrate interaction of MMPs by binding proximally to their catalytic domain. Therefore, the decreased expression of TIMPs may enhance MMP activity, thereby promoting tumor growth and invasiveness.
Importantly, we showed that menin is directly associated with the
The AR signaling axis is a critical part of tumor growth and metastatic development of PCa (Ko et al., 2015; Lin et al., 2018). However, unexpectedly, our migration assay showed that menin depletion affected the migratory potential of AR-negative DU145 and PC3 cells. At the molecular level, menin was linked to the regulation of metastasis-associated genes in DU145 cells, indicating that menin promotes metastatic behavior of tumor cells through the regulation of metastasis genes in an AR-independent manner. However, the underlying mechanisms and affected genes are expected to be distinct for each cell line. For example, genes like
In summary, our study shows an unexpected role of menin in the metastasis of PCa that does not depend on AR signaling. In light of our findings, menin may be a future target for small molecule inhibitors and act as a prognostic biomarker for PCa metastasis.
This work was supported by National Research Foundation of Korea (NRF) grants funded by the Korean government (NRF-2019R1A5A2027340, 2021R1F1A1049941, and 2019R1A2C2084716 to E.J.C. and 2012R1A3A2048767 to H.D.Y.).
T.K., K.J., E.K., K.Y., J.H.P., and J.C. performed the experiments, analyzed the data, wrote the manuscript. J.C. and J.H.K. performed bioinformatics. H.D.Y. and H.S.K. provided reagents. E.J.C. conceived the study design and wrote the manuscript. T.K. and K.J. contributed equally to this work.
The authors have no potential conflicts of interest to disclose.
Christopher J. Occhiuto, Jessica A. Moerland, Ana S. Leal, Kathleen A. Gallo, and Karen T. Liby
Mol. Cells 2023; 46(3): 176-186 https://doi.org/10.14348/molcells.2023.2191Min Ji Park, Eunji Jeong, Eun Ji Lee, Hyeon Ji Choi, Bo Hyun Moon, Keunsoo Kang, and Suhwan Chang
Mol. Cells -0001; (): https://doi.org/10.14348/molcells.2023.2174Cho-Won Kim, Hong Kyu Lee, Min-Woo Nam, Youngdong Choi, and Kyung-Chul Choi
Mol. Cells 2022; 45(12): 935-949 https://doi.org/10.14348/molcells.2022.0105