Reduced ACY1 expression correlates with poor prognosis and resistance to platinum-based chemotherapy in patients with EOC
Our previous study used gene expression microarray showed that ACY1 expression is higher in EOC tissue than in normal ovarian epithelial tissue (Figure S1A). The Gene Expression Profiling Interactive Analysis database (http://gepia.cancer-pku.cn/) also showed that the ACY1 mRNA levels were significantly higher in EOC tissue than in normal tissue (Figure S1B). Consistently, in samples from our institute (Sun Yat-sen University Cancer Center, Guangzhou, China), we found that ACY1 mRNA levels were significantly higher in EOC tissues (n = 19) than in normal tissues (n = 19) (Figure S1C). In an analysis performed using the online bioinformatics tool Kaplan-Meier Plotter (http://kmplot.com/analysis/), patients with EOC who had higher levels of ACY1 were found to have greater progression-free survival (PFS) and overall survival (OS) than those with EOC who had lower levels of ACY1 (Figure S1D).
We further determined the clinical significance of ACY1 expression by performing immunohistochemical staining of 120 archived EOC tissue specimens (Fig. 1A). As expected, patients with high levels of ACY1 (n = 59) had longer PFS (P = 0.003) and greater OS (P = 0.028) than those with low ACY1 levels (n = 61) (Fig. 1B and 1C). In addition, a multivariate analysis demonstrated that ACY1 expression was an independent prognostic factor for PFS and OS in patients with EOC (Fig. 1D and E). Furthermore, we found that elevated expression of ACY1 was inversely correlated with recurrence and a shorter progression-free interval (PFI; PFI ≤ 6 months or PFI > 6 months) (Table S1; Fig. 1F). Generally, platinum response is classified according to the PFI. The cut-off time is set at six months, defining platinum-sensitive as PFI since last line of platinum > 6 months and platinum-resistant as ≤ 6 months21. To explore the effects of ACY1 expression on response to platinum, we analyzed the correlation between ACY1 expression and platinum efficacy in 100 EOC patients treated with platinum-based chemotherapy (Figure S1E). The results showed that ACY1 levels were significantly higher in patients with EOC who were platinum-sensitive (n = 70) than in patients with EOC who were platinum-resistant (n = 30) (P < 0.01, Fig. 1G). Taken together, our results suggest that lower ACY1 levels predict resistance to platinum-based chemotherapy and poor clinical outcomes in patients with EOC.
ACY1 suppresses cisplatin resistance in EOC cells
Cisplatin is the mainstay of first-line chemotherapeutics for EOC. To further assess the effect of ACY1 on the efficacy of chemotherapy in EOC, stable ACY1-knockdown SKOV3 EOC cell lines using lentiviral short-hairpin RNA (shRNA) and ACY1-overexpressing A2780, TOV21G EOC cell lines were treated with cisplatin (Fig. 2A and S2A). A Cell Counting Kit-8 (CCK8) assay showed that ACY1 knockdown significantly increased the half-maximal inhibitory concentration (IC50) of cisplatin required to inhibit cell proliferation, whereas ACY1 overexpression decreased this IC50 (Fig. 2B–2E, S2B, and S2C). ACY1 knockdown consistently and substantially enhanced the colony-forming ability of cisplatin-treated EOC cells, whereas ACY1 overexpression inhibited this ability of cisplatin-treated EOC cells (Fig. 2F and 2G), indicating that ACY1 downregulation promoted cisplatin resistance in EOC cells. Moreover, annexin V/propidium iodide (PI) staining demonstrated that cisplatin-induced apoptosis was significantly attenuated in ACY1-knockdown cells and elevated in ACY1-overexpressing cells (Fig. 2H–2K, S2D, and S2E). Furthermore, we detected the levels of the DNA double-strand break marker gamma H2A histone family member X (γH2AX), anti-apoptotic marker B-cell leukemia/lymphoma 2 (Bcl-2), proapoptotic marker Bcl-2 associated X (Bax), and apoptotic marker cleaved poly (adenosine diphosphate-ribose) polymerase (PARP) via western blot analysis. The levels of γH2AX, Bax, and cleaved PARP were found to be reduced by ACY1 silencing but increased by ACY1 overexpression; by contrast, the level of Bcl-2 was increased by ACY1 silencing but reduced by ACY1 overexpression (Figure S2F). Taken together, the above findings indicated that ACY1 downregulation promoted cisplatin resistance in EOC cells.
To confirm the above findings in vivo, we subcutaneously inoculated three cohorts of female nude mice in the flank with ACY1-overexpressing, ACY1-knockdown, or control TOV21G cells, respectively. When the tumor volume of each cohort reached 100–150 cm3, the mice were treated with either cisplatin or saline solution. Cisplatin treatment reduced the tumor volume and weight in the ACY1-overexpressing cohort significantly more than in the ACY1-knockdown cohort (Fig. 2L–N), indicating that ACY1 upregulation attenuated cisplatin resistance in vivo. Overall, our results provide evidence that ACY1 depletion induces resistance to cisplatin in EOC cells.
ACY1 physically interacts with GSTP1
ACY1 is a cytosolic metalloenzyme that typically exerts its effects via direct interactions with proteins7. To characterize the molecular mechanisms underlying the roles of ACY1 in the resistance of EOC to cisplatin, we transfected an ACY1 plasmid into HEK-293T cells and performed mass spectrometry (MS) to identify ACY1-binding partners. There were several top potential ACY1-binding partners, such as RPS9, KRT1 and KRT9 (Fig. 3A and 3B; Table S2), with GSTP1 being chosen for further analysis owing to its known roles in drug metabolism and preventing oxidative damage. Moreover, GSTP1 upregulation markedly increased the resistance of patients with EOC to platinum-based chemotherapeutics.19. The results of the co-immunoprecipitation (co-IP) assay showed that endogenous ACY1 and GSTP1 co-precipitated in A2780 and TOV21G cells (Fig. 3C). Exogenous ACY1 was reciprocally co-immunoprecipitated with GSTP1 in HEK-293T cells transfected with Flag-ACY1 and HA-GSTP1 plasmids (Fig. 3D). Furthermore, GST pull-down assay identified that GSTP1 directly bound to ACY1 in vitro (Fig. 3E), and additional immunofluorescence assays revealed positive signals of ACY1 and GSTP1 colocalization in the cytoplasm of EOC cells (Fig. 3F). To determine which domain of ACY1 mediates the interactions between ACY1 with GSTP1, we constructed a series of deletion-mutant plasmids of Flag-ACY1 (Fig. 3G). Reciprocal co-IP assays revealed that the C-terminal M20 peptidase domain of ACY1 interacted with GSTP1 (Fig. 3H). Based on the above observations, we conclude that ACY1 physically interactes with GSTP1.
ACY1 decreases GSTP1 stability by facilitating FBX8-mediated ubiquitination and degradation of GSTP1
To explore whether physical interactions between GSTP1 and ACY1 regulate the expression of GSTP1, we quantified the levels of GSTP1 in the same cohort of EOC tissue samples used for analyzing the levels of ACY1 with IHC staining. This revealed that ACY1 levels were negatively correlated with GSTP1 levels (R = − 0.294, P = 0.0012) (Fig. 4A and 4B). Western blot analysis showed that ACY1 knockdown increased the levels of GSTP1 protein, whereas ACY1 overexpression decreased the levels of GSTP1 protein (Fig. 4C). Levels of ectopic HA-GSTP1 were also decreased in ACY1-overexpressing cells but increased in ACY1-silenced EOC cells (Fig. 4D and S3A). However, the expression of ACY1 did not significantly affect the levels of GSTP1 mRNA (Figure S3B and S3C), suggesting that ACY1 may regulate the translation of GSTP1 mRNA into protein or the stability of the protein.
To better understand the mechanisms via which ACY1 regulates GSTP1 protein levels, a cycloheximide (CHX) chase assay was conducted to assess GSTP1 protein half-life. GSTP1 protein half-life was found to be shortened by ectopic ACY1 (Fig. 4E and 4F) but lengthened by ACY1 silencing (Fig. 4G and 4H). Furthermore, GSTP1 protein levels were elevated in ACY1-overexpressing cells treated with the proteasome inhibitor MG132 (Fig. 4I and 4J). Similarly, the level of GSTP1 ubiquitination was enhanced by ACY1 overexpression (Fig. 4K) but reduced by ACY1 knockdown (Fig. 4L). Taken together, these results indicated that ACY1 attenuated the stability of GSTP1 via the ubiquitin-proteasome pathway.
F-box only protein 8 (FBX8), an E3 ubiquitin ligase, induces the degradation of GSTP1 by ubiquitin and to suppress CRC progression22. To determine whether ACY1 reduces the stability of GSTP1 via FBX8-mediated ubiquitination, we conducted a co-IP and western blot assay. This demonstrated that the ability of ACY1 to enhance GSTP1 ubiquitination was significantly inhibited in FBX8-silenced cells (Fig. 4M). Overall, these findings show that ACY1 promotes the ubiquitin–proteasome-dependent degradation of GSTP1 via the E3 ligase activity of FBX8.
ACY1 reduces GSTP1 stability and thereby increases intracellular ROS levels and inhibits DNA damage repair in cisplatin-treated EOC cells
To evaluate whether ACY1 suppresses cisplatin resistance through the action of GSTP1, we used small interfering RNA (siRNA) to downregulate the expression of GSTP1 in ACY1-knockdown cells. CCK8 and annexin V/PI assays revealed that GSTP1 knockdown significantly abolished the enhanced growth and survival capacities of cisplatin-treated EOC cells induced by ACY1-silencing (Fig. 5A–5D).
GSTP1 protects cancer cells from DNA damage and induced drug resistance by activating the ATM–CHK2–p53 signaling pathway17. Thus, to determine whether ACY1 regulates cell response to cisplatin via the anti-oxidation and DNA damage repair pathway activation effects of GSTP1, we measured the levels of intracellular ROS and related checkpoint kinases in the DNA damage repair signaling pathway, including ATM, phosphorylated ATM, CHK2, and phosphorylated CHK2. The results of flow cytometry assays revealed that ACY1 depletion decreased the intracellular levels of ROS in cisplatin-treated SKOV3 cells (Fig. 5E and 5F), whereas ACY1 overexpression increased the levels of ROS in cisplatin-treated A2780 and TOV21G cells (Fig. 5G and 5H). Moreover, the results of western blots revealed that the levels of phosphorylated ATM and CHK2 were increased upon ACY1 knockdown and decreased upon ACY1 overexpression (Fig. 5I). In sum, these findings show that ACY1 suppresses cisplatin resistance in EOC cells by blocking GSTP1-mediated regulation of intracellular ROS levels and activation of the ATM–CHK2 signaling pathway
ACY1 deacetylates GSTP1 at K30, which reduces GSTP1 stability
ACY1 deacetylates α-acetylated amino acids in the N-terminal peptide of intracellular proteins11, and lysine (K) acetylation is the most common type of acetylation and can prevent ubiquitination and proteasome-dependent protein degradation23. Thus, we realized it was important to assess whether ACY1 regulates GSTP1 acetylation. We therefore evaluated the effect of ACY1 expression on GSTP1 acetylation using a pan-acetylated K antibody. The K acetylation of ectopic HA-GSTP1 was increased in ACY1-knockdown SKOV3 cells (Fig. 6A, left panel) but decreased in ACY1-overexpressing A2780, HEK-293T, and TOV21G cells (Fig. 6A, right panel; S4A), implying that ACY1 may be responsible for GSTP1 deacetylation. To identify the specific acetylation sites of GSTP1, we isolated and purified GSTP1 from HEK-293T cells transfected with HA-GSTP1. MS analysis revealed that two K sites––K30 and K45––on the N-terminal GST domain of this purified GSTP1 were acetylated (Fig. 6B and 6C), which has not previously been reported. We therefore mutated both K30 and K45 to arginine (R) (K30R and K45R; acetylation-dead mutant), which resulted in significantly reduced the levels of total and acetylated ectopic GSTP1 protein in EOC cells (Fig. 6D). Moreover, a CHX chase assay showed that GSTP1-K30R had a shorter half-life than GSTP1-WT and GSTP1-K45R in EOC cells (Fig. 6E and 6F). Furthermore, the levels of ubiquitinated GSTP1-K30R were higher than the levels of ubiquitinated GSTP-WT and GSTP1-K45R (Fig. 6G), suggesting that K30 was the acetylation site that mediated GSTP1 ubiquitination. To confirm that ACY1 impaired GSTP1 stability via deacetylation at K30, we measured the levels of ubiquitinated ectopic HA-GSTP1 in HEK-293T cells transfected with GSTP1-acetylated site mutant plasmids and ACY1 siRNA. The results showed that ACY1 knockdown significantly reduced the levels of ubiquitinated GSTP1-WT and GSTP1-K45R but did not inhibit the K30 acetylation-mediated increase in GSTP1 ubiquitination (Fig. 6H). Moreover, CCK8 and annexin V/PI assays revealed that GSTP1-WT but not GSTP1-K30R significantly rescued the reduced growth and survival capacities of cisplatin-treated ACY1-overexpressing EOC (Fig. 6I–6L). Overall, our results demonstrate that ACY1 deacetylates GSTP1 at K30 to reduce GSTP1 stability in EOC cells, which leads to GSTP1 degradation via the ubiquitin–proteasome pathway.
HDAC4-mediated suppression of H3K27Ac inhibits ACY1 transcription in cisplatin-treated EOC cells
Owing to the lack of agonists that can bind ACY1, we explored the upstream mechanism underlying ACY1 expression in EOC cells to extend our study findings to a clinical setting. As histone acetylation plays an important role in chemoresistance mechanisms in EOC24, we first evaluated modifications of the ACY1 promoter recorded on the UCSC Genome Browser (http://genome.ucsc.edu/). As shown in Fig. 7A, there was substantial H3K27Ac in the ACY1 promoter region, implying that ACY1 expression is regulated by chromatin acetylation. HDACs eliminate acetyl groups from histones, resulting in reduced gene expression. According to the LinkedOmics portal (http://linkedomics.org/), among all HDACs, only the mRNA levels of HDAC4 are negatively correlated with ACY1 mRNA levels in OC (R = − 0.229, P = 0.0390) (Fig. 7B and S5A). In addition, Stronach et al. reported that HDAC4 is significantly upregulated in clinical samples following acquired platinum resistant relapse and promotes platinum resistance in OC cells25. Therefore, we speculated that HDAC4 regulates the expression of ACY1 via an epigenetic modification in EOC upon treatment of platinum-based chemotherapy.
Next, we measured the levels of ACY1 and HDAC4 in cisplatin-treated SKOV3, A2780 and TOV21G cells at various time points. This revealed that levels of ACY1 reduced gradually, whereas those of HDAC4 increased gradually upon cisplatin treatment at increasing time (Fig. 7C), implying that cisplatin induced HDAC4 expression but inhibited ACY1 expression. To confirm whether HDAC4 directly regulated ACY1 expression, we examined the levels of ACY1 mRNA and protein in EOC cell lines using two specific siRNAs targeting HDAC4. We found that HDAC4 knockdown significantly increased the levels of ACY1 mRNA and protein (Fig. 7D, 7E, and S5B). Moreover, a chromatin immunoprecipitation (ChIP) assay indicated that the ACY1 promoter region was enriched in H3K27Ac signals, and that this enrichment was increased by HDAC4 knockdown (Fig. 7F). Next, we performed CCK8 and annexin V/PI assays, which demonstrated that HDAC4 knockdown significantly attenuated the promotive effect of ACY1 silencing on the growth and survival capacities of cisplatin-treated EOC cells (Fig. 7G–7J). These data suggest that HDAC4-mediated suppression of H3K27Ac on the ACY1 promoter at least partly accounts for cisplatin treatment of EOC cells triggering the repression of ACY1 transcription, which leads to these cells exhibiting cisplatin resistance.