PLXND1 is upregulated in NEPC and prostate cancer cells showing neural lineage plasticity.
Analysis of publicly available RNA-seq datasets revealed upregulated mRNA levels of PLXND1 and neuroendocrine feature genes (CHGA, NCAM1, SYP, and ENO2), along with downregulated AR and its targeting genes (KLK2, KLK3, NKX3-1, TMPRSS2, FKBP4, and FKBP5) in NEPC tumors compared to CRPC (Beltran 2016, GSE160393) (Fig. 1A, Fig.S1D) [20], which was observed in both AMPC (amphicrine expression of both AR and neuroendocrine markers) and NEPC, compared to CRPC, castration-sensitive prostate cancer (CSPC), and DNPC (double-negative expression of AR and neuroendocrine markers) (GSE160393, GSE126078) (Fig. 1B-1C) [24, 25]. Additionally, HuPSA (Human Prostate Single cell Atlas) data analysis demonstrated higher PLXND1 mRNA levels in NEPC patients by considering the heterogeneous populations of prostate cancer cells (Fig.S1A). To investigate whether enzalutamide treatment affects PLXND1 expression, data analysis showed that PLXND1 was upregulated in enzalutamide-treated prostate cancer cells compared to the DMSO treatment group (GSE215653) (Fig.S1B) [26]. Furthermore, our RNA-seq data revealed upregulation of both PLXND1 and neuroendocrine markers, such as ENO2, CHGA, NCAM1, and SYP, in C4-2B-MDVR cells compared with parental C4-2B cells (Fig.S1E). GSEA analysis indicated a significant enrichment of neural lineage pathways in enzalutamide-treated prostate cancer cells (GSE215653 and GSE 151083) (Fig.S1F-S1G) [26, 27], including pathways related to synapse assembly, synaptic membrane, neuron projection terminus, synaptic transmission glutamatergic, neurotransmitter receptor activity, and distal axons. To assess the association between PLXND1 expression levels and prostate cancer patient prognosis, Kaplan-Meier survival analysis demonstrated that patients with higher PLXND1 expression had inferior survival rates compared to those with lower PLXND1 expression (TCGA: HR = 1.80 (1.19–2.75), P = 0.006; GSE21032: HR = 3.23 (1.67–6.24), P < 0.001) [22, 28]. Correlation analysis revealed a positive correlation between PLXND1 expression and neuroendocrine features (Beltran 2016: CHGA (r = 0.42, P = 0.002), ENO2 (r = 0.53, P < 0.0001), SYP (r = 0.59, P < 0.0001), and NCAM1 (r = 0.32, P = 0.02; GSE126078: CHGA (r = 0.59, P < 0.0001), ENO2 (r = 0.60, P < 0.0001), SYP (r = 0.43, P < 0.0001), and NCAM1 (r = 0.55, P < 0.0001)) (Fig. 1F and Fig.S1H) [20, 25]. We measured PLXND1 expression and associated neuroendocrine characteristics across different human prostate cancer cell lines, patient-derived xenograft (PDX) tumors, and prostate cancer patient samples. The results showed that PLXND1 was negative in C4-2B cells but was highly expressed in NE-like or NEPC cell lines (C4-2B-MDVR, CWR22Rv1, and H660) (Fig. 1G-1H). Further investigation revealed upregulated PLXND1 expression in C4-2B-2M (enzalutamide treatment for 2 months) and C4-2B-MDVR cells compared to that in parental C4-2B cells (Fig. 1I). Additionally, PLXND1 was more highly expressed in NEPC PDX tumors (LuCaP 49, LuCaP 93, LuCaP 145.2, and LuCaP 173.1) than in CRPC PDX tumors (UCD1172, UCD1173, UCD1178, and LuCaP35CR) (Fig. 1J). Immunohistochemistry (IHC) staining confirmed high PLXND1 expression in NEPC tumors (C4-2B-MDVR and LuCaP 93) compared to that in C4-2B tumors (Fig. 1K). Importantly, we screened 46 CRPC and three NEPC patient samples and found that PLXND1 staining was significantly increased in the NEPC patient samples (Fig. 1L). In summary, the data suggest that PLXND1 is upregulated in NEPC and cells gaining neural lineage plasticity features.
AR signaling negatively regulates the expression of PLXND1 in enzalutamide-resistant prostate cancer.
To examine whether AR signaling regulates the expression of PLXND1, PLXND1 protein expression was determined in C4-2B-MDVR cells maintained in charcoal-stripped fetal bovine serum (CS-FBS) and regular FBS conditions. PLXND1 expression was significantly upregulated in CS-FBS compared to FBS (Fig. 2A). Both RNA-seq and GEO data (GSE52169) revealed that dihydrotestosterone (DHT) treatment significantly downregulated the mRNA levels of PLXND1 in prostate cancer (Fig.S2A-S2B) [29]. Further investigation confirmed that DHT treatment significantly suppressed the mRNA and protein expression of PLXND1 in a dose- and time-dependent manner in C4-2B-MDVR cells (Fig. 2B-2E). Additionally, DHT and enzalutamide combination treatment showed that enzalutamide could rescue both PLXND1 mRNA and protein expression inhibited by DHT in C4-2B-MDVR cells (Fig. 2F-2G). To determine whether the AR protein regulates PLXND1 expression, full-length AR (AR-FL) was knocked down with siRNA in C4-2B-MDVR cells. As shown in Fig. 2H-2I and Fig.S2C, AR-FL knockdown significantly increased the expression of PLXND1 mRNA and protein. Furthermore, DHT significantly reduced PLXND1 expression, and AR-FL knockdown blocked the inhibitory effect of DHT on PLXND1 expression (Fig. 2I, Fig.S2C). This was also confirmed by analyzing prostate cancer patient cohorts (Fig. 2J-2K, Fig.S2D-S2E). AR and its downstream targeting genes, such as KLK2, KLK3, and NKX3-1, were negatively correlated with PLXND1 in Beltran 2016, GSE126078, SU2C/PCF, and MSK 2022 databases [20, 21, 25]. In summary, these data suggest that AR signaling negatively regulates PLXND1 expression in enzalutamide-resistant prostate cancer cells.
Knockdown of PLXND1 represses the cell proliferation and improves enzalutamide treatment.
To evaluate whether PLXND1 plays a role in regulating the aggressive characteristics of neuroendocrine prostate cancer (NEPC) cells, we employed two distinct siRNAs to knockdown PLXND1 in C4-2B-MDVR, CWR22Rv1, and H660 cells, all of which exhibit neuroendocrine traits [30–32]. The knockdown effect was validated using RT-qPCR (Fig.S3A). Subsequently, we observed that PLXND1 knockdown suppressed cell proliferation and colony formation in C4-2B-MDVR cells compared with the control groups. Similar inhibitory effects were observed in CWR22Rv1 and H660 cells (Fig. 3A-3B). Additionally, PLXND1 knockdown led to reduced expression of CDK2, CyclinA, CyclinD1, and CyclinE, along with increased expression of cleaved-PARP in C4-2B-MDVR, CWR22Rv1, and H660 cells (Fig. 3C). Importantly, we found that PLXND1 knockdown significantly improved enzalutamide treatment of C4-2B MDVR and CWR22Rv1 cells (Fig. 3D and Fig.S3B). Moreover, we assessed the effect of PLXND1 knockdown in an H660 organoid model. The results demonstrated that Silencing PLXND1 expression via siRNA significantly inhibited the viability and growth of H660 organoids (Fig. 3E). In summary, these findings suggest that PLXND1 could potentially serve as a therapeutic target for NEPC, given its involvement in regulating key cellular processes and aggressive behavior of NEPC cells.
Knockdown of PLXND1 decreases the neuroendocrine traits.
To evaluate the impact of PLXND1 knockdown on neuroendocrine traits and associated pathways, we conducted RNA-seq analysis of C4-2B-MDVR and H660 cells with silenced PLXND1. The results revealed that 287 genes were significantly downregulated and 349 genes were significantly upregulated in C4-2B-MDVR cells (Fig. 4A). Similarly, in H660 cells, 431 genes were significantly downregulated and 1008 genes were significantly upregulated (Fig. 4B). Subsequent analysis showed that six neural lineage pathways were downregulated in C4-2B-MDVR cells with PLXND1 knockdown compared to the control, including the negative regulation of axonogenesis, developmental growth, synapse assembly, neuron differentiation, neuron projection development, and neurogenesis (Fig. 4C). GO analysis also revealed the downregulation of neural lineage pathways in H660 cells transfected with siPLXND1 (Fig.S4A). Further GOBP analysis indicated that five pathways were upregulated in C4-2B-MDVR cells with PLXND1 knockdown compared to the control, including regulation of the apoptotic signaling pathway, signal transduction by P53 class mediator, regulation of the extrinsic apoptotic signaling pathway, positive regulation of the apoptotic signaling pathway, and negative regulation of the cell cycle G1/S phase transition. KEGG analysis identified upregulation of the P53 signaling pathway and apoptosis in C4-2B-MDVR cells with PLXND1 knockdown compared with the control. Reactome analysis further revealed that four pathways regulating the cell cycle and apoptosis were significantly upregulated in C4-2B-MDVR cells with PLXND1 knockdown (Fig. 4D). GSEA analysis demonstrated that four neural lineage pathways were downregulated in C4-2B-MDVR cells with PLXND1 knockdown, including neurotransmitter uptake, regulation of receptor localization to synapses, synaptic transmission GABAergic, and regulation of GABAergic synaptic transmission (Fig. 4E). Apoptosis pathways were also upregulated in H660 cells transfected with siPLXND1 (Fig.S4B). Additionally, PLXND1 knockdown downregulated the protein expression of neuroendocrine markers, including CHGA, NSE, and SYP (Fig. 4F). Finally, heatmap analysis illustrated the downregulation of neural lineage pathway genes and the upregulation of apoptosis pathway genes in C4-2B-MDVR cells transfected with siPLXND1 (Fig. 4G). In summary, our data suggest that PLXND1 knockdown decreases neuroendocrine traits of NEPC cells.
Knockout of PLXND1 by CRISPR/Cas9 inhibits the NEPC cells in vitro, organoids viability, and tumor growth in vivo.
Subsequently, LentiCRISPR v2 plasmids were used to knock out PLXND1 in the C4-2B-MDVR, CWR22Rv1, and H660 cells. The knockout effect was validated by western blotting in C4-2B-MDVR, CWR22Rv1, and H660 cells (Fig. 5A). The results indicated that PLXND1 knockout suppressed proliferation and colony formation in C4-2B-MDVR cells compared to the control (Fig. 5B-5C), as well as in CWR22Rv1 (Fig.S5A-S5B) and H660 cells (Fig. 5D). Moreover, we assessed the effects of PLXND1 knockout in LuCaP49 and LuCaP93 organoid models and demonstrated that CRISPR/Cas9-mediated PLXND1 knockout significantly inhibited the viability and growth of LuCaP49 and LuCaP93 organoids (Fig. 5E, Fig.S5C). To determine whether PLXND1 knockout inhibited NEPC tumor growth in vivo, CWR22Rv1 cells, including sgControl, sgPLXND1#1, and sgPLXND1#2, were injected to establish xenografts. PLXND1 knockout significantly inhibited tumor growth in CWR22Rv1 xenografts (Fig. 5F-5H). IHC staining indicated that Ki67 expression was also suppressed in the sgPLXND1 group compared with that in the sgControl group (Fig. 5I). Furthermore, compared with the sgControl group, CDK2 and CDK4, along with their related cyclins, such as CyclinA, CyclinD1, and CyclinE, were significantly suppressed in the sgPLXND1 groups (Fig. 5J). In summary, these data confirm that PLXND1 affects NEPC proliferation and could serve as a potential therapeutic target for NEPC.
HSP70 inhibition affects the protein stability of PLXND1.
For a protein to become functional, it requires correct folding and assembly, a process that relies on the chaperone HSP70. To determine whether HSP70 binds to PLXND1 and regulates PLXND1 expression, we conducted co-IP assays in HEK293 and CWR22Rv1 cells. The results indicated that HSP70 binds to PLXND1 in both HEK293 and CWR22Rv1 cells (Fig. 6A-6B). Subsequently, we knocked down HSP70 in CWR22Rv1 cells using siRNA, and RT-PCR and western blotting were employed to validate the knockdown efficacy (Fig. 6C-6D). The findings revealed that HSP70 knockdown downregulated the protein expression of PLXND1 without affecting its mRNA expression of PLXND1 (Fig. 6C-6D). Using the HSP70 allosteric inhibitor JG231, we found that JG231 treatment significantly reduced the expression of PLXND1, CDK2, CyclinA, CyclinD1, CyclinE, and neuroendocrine markers (CHGA, NSE, and SYP) and increased the expression of cleaved-PARP and cleaved-Caspase7 in C4-2B-MDVR, CWR22Rv1, and H660 cells (Fig. 6E, Fig.S6A). To determine whether the decrease in PLXND1 protein expression induced by JG231 treatment was mediated through the proteasome pathway, we added the proteasome inhibitor MG132 to CWR22Rv1 cells. Although JG231 reduced PLXND1 protein expression, the addition of MG132 blunted the effects of JG231 (Fig. 6F). Furthermore, we investigated whether JG231 affects PLXND1 protein stability in CWR22Rv1 cells using the cycloheximide (CHX) chase assay and found that JG231 treatment significantly shortened the half-life of PLXND1 (Fig. 2G). Notably, JG231 treatment inhibited the growth of LuCaP49 and LuCaP93 NEPC organoids in a dose-dependent manner (Fig. 6H, Fig.S6B). Collectively, these data suggest that the chaperone protein HSP70 may control the turnover of PLXND1, and HSP70 inhibition may indirectly target PLXND1 in NEPC.