Zhang et al. J Transl Med (2021) 19:98 https://doi.org/10.1186/s12967-021-02770-0 Journal of Translational Medicine

RESEARCH Open Access Hypermethylation of 2 promotes colorectal cancer proliferation and is associated with poor prognosis Hui Zhang1†, Chenxin Xu2†, Chen Shi2, Junying Zhang2, Ting Qian3, Zhuo Wang2, Rong Ma2, Jianzhong Wu2, Feng Jiang4* and Jifeng Feng2*

Abstract Background: The epigenetic abnormality of tumor-associated contributes to the pathogenesis of colorectal carcinoma (CRC). However, methylation in colorectal cancer is still poorly characterized. Method: By integration of DNA methylation data from the GEO database and expression data from The Cancer Genome Atlas database, the aberrantly methylated genes involved in CRC tumorigenesis were identifed. Subsequent in vitro experiments further validated their role in CRC. Results: We performed integrative genomic analysis and identifed HPSE2, a novel tumor suppressor gene that is frequently inactivated through promoter methylation in CRC. K-M survival analysis showed that hypermethylation– low expression of heparanase 2 (HPSE2) was related to poor patient prognosis. Overexpression of HPSE2 reduced cell proliferation in vivo and in vitro. HPSE2 could regulate the p53 signaling pathway to block the cell cycle in G1 phase. Conclusion: HPSE2, a novel tumor suppressor gene that is frequently inactivated through promoter methylation in CRC. HPSE2 performs a tumor suppressive function by activating the p53/ p21 signaling cascade. The promoter hypermethylation of HPSE2 is a potential therapeutic target in patients with CRC, especially those with late-stage CRC. Keywords: Colorectal cancer, Heparanase 2, Methylation, Biomarker, Proliferation

Background incidence and mortality among people aged 50 and older Colorectal cancer (CRC) remains the third most com- in recent years [2]. mon cancer and the second leading cause of cancer Te epigenetic abnormality of tumor pathogenic genes, deaths worldwide [1] despite dramatic drops in its overall particularly the DNA methylation of selected gene pro- moters, contributes to the pathogenesis of CRC [3, 4]. DNA methylation is a covalent DNA modifcation that converts the DNA base cytosine to 5-methylcytosine; *Correspondence: [email protected]; [email protected] this process is catalyzed by DNA methyltransferases [5]. †Hui Zhang and Chenxin Xu contributed equally to this work 2 Research Center for Clinical Oncology, Jiangsu Cancer Hospital, Jiangsu Under physiological conditions, CpG island methyla- Institute of Cancer Research, The Afliated Cancer Hospital of Nanjing tion actively regulates the balance between DNA meth- Medical University, 42 Baiziting, Nanjing 210000, Jiangsu, People’s ylation and DNA demethylation to maintain proper gene Republic of China 4 Jiangsu Key Laboratory of Molecular and Translational Cancer Research, expression patterns [6]. Aberrantly methylated genes can Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The promote the pathogenesis of CRC by regulating specifc Afliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, signaling pathways [7]. CpG island methylator phenotype Nanjing 210000, Jiangsu, People’s Republic of China Full list of author information is available at the end of the article (CIMP), a specifc pattern of promoter methylation, may

© The Author(s) 2021. This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, 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 changes were made. 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://creativeco​ ​ mmons.org/licen​ ses/by/4.0/​ . The Creative Commons Public Domain Dedication waiver (http://creativeco​ mmons​ .org/publi​ cdoma​ in/​ zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data. Zhang et al. J Transl Med (2021) 19:98 Page 2 of 13

serve as a biomarker for early detection and as a tool for stage and M stage) and HPSE2 on the OS of patients with monitoring patients with CRC. DNA methylation altera- CRC. Te HR and 95% confdence interval were assessed. tions may also be useful tools for CRC diagnosis, predic- Multivariate Cox regression analysis was used to verify tion, and treatment [8, 9]. However, methylation in CRC the independent predictive capacity of HPSE2 when remains poorly characterized. Te development of Te compared with that of other clinical factors. Cancer Genome Atlas Network (TCGA dataset) and other public databases has enabled comprehensively ana- CpG island methylation of HPSE2 lyzing the molecular characteristics of cancer genomes, Te methylation data for the CpG islands of the HPSE2 transcriptomes, epigenomics, and proteomes, thus open- promoter, which are located 3 kb upstream, were down- ing up a new path for cancer molecular diagnosis [10, 11]. loaded from TCGA database. We obtained 49 CpG We previously screened two tumor suppressor genes islands and analyzed their correlation with gene expres- with known functions that are regulated by methyla- sion. Cor < − 0.3 and P < 0.05 was considered statistically tion, which confrmed the reliability of our analysis [12]. signifcant and retained for further analysis. To fnd novel pathogenic genes, we analyzed and identi- fed 10 aberrantly methylated genes by integrating DNA Sample collection methylation data from the GEO database and gene Sixty patients with pathologically confrmed CRC diag- expression data from TCGA database. On the basis of nosed within the past one year at the Afliated Cancer subsequent Kaplan–Meier plot validation, we focused Hospital of Nanjing Medical University (Nanjing, China) on heparanase 2 (HPSE2) whose hypermethylation– were enrolled in the study. Peripheral blood was obtained low expression was related to poor patient prognosis. preoperatively from 45 patients with CRC. In addition, HPSE2 overexpression reduced cell proliferation in vivo the peripheral blood samples of 44 healthy controls were and in vitro. HPSE2 could regulate the p53/P21 sign- collected from the Afliated Geriatric Hospital of Nan- aling pathway to arrest the cell cycle in the G1 phase. jing Medical University in Nanjing, China. None of the Collectively, we performed integrative genomic analy- patients received preoperative chemotherapy or radio- sis and identifed HPSE2 as a tumor suppressor gene in therapy. All the specimens were immediately frozen CRC. Aberrantly expressed HPSE2 could promote tumor in tubes containing RNAlater preservation liquid after aggressiveness via the p53/p21 signaling pathway. removal and stored at liquid nitrogen. Tis study was approved by the Ethics Boards of Jiangsu Cancer Hospi- Methods tal. Written informed consent was signed by each patient. Screening of methylation‑regulated genes from the GEO and TCGA databases Cell culture, stable cell line construction, To screen for methylation-regulated genes, we counted and HPSE2‑overexpression and analyzed aberrantly methylated genes between Human CRC cell lines SW480, HCT116, SW620, LOVO, tumors and adjacent normal tissues in GSE17648 and DLD1, and NCM460 were purchased form the Ameri- GSE29490 from the GEO database (https​://www.ncbi. can Type Culture Collection (Manassas, Virginia, USA). nlm.nih.gov/geo/). P < 0.05 and | logFC |≥ 1 were consid- Te cells were cultured at 37 °C in a 5% ­CO2 incubator ered statistically signifcant. Correspondingly, genes that in Dulbecco’s modifed Eagle’s medium (KeyGEN Bio- were abnormally expressed in CRC were screened from TECH, Jiangsu, China) with 10% fetal bovine serum TCGA database (https​://porta​l.gdc.cance​r.gov/) (P < 0.05 (Gibco, USA). Lentivirus-HPSE2 (LV-HPSE2) were con- and |log FC |≥ 3). By taking the intersection, we obtained structed in the lentivirus vector GV492 by a commercial genes showing hypermethylation–low expression and service (Genechem Biotech Inc, Shanghai, China), while hypomethylation–high expression in tumor tissues. lentivirus vector GV492 was adopted as the control. LV- We performed Pearson correlation (cor) to evaluate the HPSE2 was packaged in 293 T cells. Te supernatant of relationship between gene expression and methylation. cell culture medium containing lentivirus granules was Cor < − 0.3 and P < 0.05 were considered signifcant and collected and the viral titer of the virus solution was further analyzed. Kaplan–Meier plot survival analysis determined. HCT116 and SW480 with a growth fusion was conducted to evaluate the role of methylation-regu- degree of about 80% were digested by trypsinase and the lated genes in the prognosis of patients with CRC. cell suspension was prepared. Cells were seeded into six- 6 well plates at 1.0 × ­10 cells per well and maintained at Univariate and multivariate Cox regression analysis 37℃ in a humidifed atmosphere of 5% ­CO2. Cells were Univariate and multivariate Cox proportional hazard divided into two groups, one group was added with 10 ml regression analyses were performed to investigate the virus solution and the other group was added with 10 ml efects of various clinical features (age, gender, T stage, N empty vector virus solution. Te multiplicity of infection Zhang et al. J Transl Med (2021) 19:98 Page 3 of 13

­(MOIHCT116 and MOI­ SW480) was 10. After 16 h of cell were added to the culture for 1 h in the dark to detect culture, the culture medium was replaced. After 3 days absorbance at 450 mm. of infection, the cells were in good condition. Te posi- tive clones were screened by using a complete culture Transwell experiments medium containing 2.0 mg/mL puromycin for 4 weeks. A total of 200 µl of cell suspension was diluted with serum-free medium and added to the upper culture dish RNA extraction, reverse transcription and qRT‑PCR of a Transwell chamber (8 mm well, Corning, USA) to Tissue and blood RNA were extracted according to the ensure that the number of cells in the upper layer was not 4 instruction of Tissue RNA Kit (OMEGA bio-tek, R6688- less than 5 × ­10 . A total of 500 µl of medium containing 01, USA) and Blood RNA Kit (OMEGA bio-tek, R6814- 10% FBS was added to the lower culture dish. After 36 h 01C, USA). TRIzol reagent (Invitrogen) was utilized to of incubation at room temperature, the cell membrane extract RNAs from cultured cells according to manu- was fxed with 4% paraformaldehyde, stained with crystal facturer’s instructions. A ratio of (A260)/(A280) is an violet, and photographed under an inverted microscope. indication of nucleic acid purity. A value greater than 1.8 indicates > 90% nucleic acid purity. For qRT-PCR, Cell cycle experiments 1 μg RNAs were inversely transcribed into 20 μl cDNA Cell counting experiments and cell cycle experiments with a Reverse Transcription Kit (Takara, Dalian, China). were performed by using a cell cycle detection kit (Key- qRT-PCR was analyzed according to our previously pub- GEN BioTECH #KGA511-KGA512) in accordance with lished article [12]. Te relative expression of HPSE2 was instructions. performed for three independent times and normalized using the ­2− ΔΔCt method relative to GAPDH. Xenograft mouse carcinogenesis model Te primers were as follows: GAPDH-Forward: GGT​ All animal research procedures were approved by the GAA​GGT​CGG​AGT​CAA​CG, Institutional Animal Care and Use Committee of Sun GAPDH-Reverse: TGG​GTG​GAA​TCA​TAT​TGG​ Yat-sen University. Twelve female 6-week-old BALB/ AACA, cA-nude mice were purchased from Nanjing (Nanjing, 6 HPSE2-Forward: ATG​GCC​GGG​CAG​TAA​ATG​G, China). A total of 1 × ­10 HCT116 cells were injected HPSE2-Reverse: GCT​GGC​TCT​GGA​ATA​AAT​CCG. subcutaneously into the fanks of the mice. Te weight of the mice was measured every week, and the size of sub- Western blot analysis cutaneous tumors was observed. Te animals were euth- Total protein was lysed with RIPA extraction reagent anized after 5 weeks, and tumor volume was measured 3 (TermoFisher, USA) that contained with a protease (volume = 4/3pr ). Finally, tumor tissues were embedded, inhibitor cocktail (Beyotime Biotechnology, China). Pro- fxed, and prepared for IHC staining. Cancer tissue was tein lysates were separated by 10% sodium dodecyl sul- cut into 6.0 mm sections and stained with anti-ki67 anti- fate polyacrylamide gel electrophoresis (SDSP-AGE) and body for IHC. Images were captured by using an AxioVi- transferred to 0.22 mm polyvinylidene fuoride mem- sion Rel. 4.6 computer image analysis system (Carl Zeiss). branes (Millipore, USA). Te membranes were subse- quently blocked in 5% defatted milk and incubated with GSEA enrichment analysis primary antibodies overnight at 4 °C. Specifc bands were We performed GSEA enrichment analysis with GSEA visualized by ECL chromogenic substrate and quanti- v4.0.3 (https​://www.gsea-msigd​b.org/gsea/downl​oads. fed by densitometry (Quantity One software, BioRad). jsp). Te reference gene set of KEGG enrichment analy- Te following antibodies were used: HPSE2 (1:500; sis was c2.cp.kegg.v7.2.symbols.gmt (http://www.gsea- #ab127204, abcam, Cambridge, UK), p53 (1:1000; #2527, msigd​b.org/gsea/downl​oads.jsp). Gene expression data Cell Signaling Technology, Inc., MA, USA), p21 (1:1000; of 647 CRC samples were downloaded from TCGA data- #2947, Cell Signaling Technology, Inc., MA, USA), base and classifed into 2 groups (High HPSE2 expression GAPDH (1:1000; #5174, Cell Signaling Technology, Inc., group vs. Low HPSE2 expression group) by the median MA, USA), and ki67 (1:1000; #ab15580, abcam, Cam- expression of HPSE2. Ten, the rest steps were carried bridge, UK). out according to the conventional analysis method of GSEA software [13, 14]. CCK8 Cell proliferation was evaluated by using a CCK-8 kit Statistical analysis (Dojindo, Japan) in accordance with the manufacturer’s RNA-seq data analysis was performed with R 3.6.0 soft- 3 instructions. Briefy, 3 × ­10 cells were seeded in a 96-well ware. Te R packages “limma”, “survival”, “plyr”, “ggplot2”, plate, and 10 µl of CCK8 solution and 100 µl of DMEM “grid”, “gridExtra”, and “ggpubr” were used according to the Zhang et al. J Transl Med (2021) 19:98 Page 4 of 13

instruction of bioconductor website (http://www.bioco​ USA) was applied to statistically analyze qPCR results. nductor.org/​ ). All of the measurement data were expressed Student’s t-test was used to evaluate statistical diferences as mean ± SD. Univariate and multivariate Cox regression between 2 groups. P < 0.05 was considered statistically analysis were performed with SPSS 22 software (Chicago, signifcant. IL, USA). GraphPad Prim 5 (GraphPad Software, La Jolla,

abc

d

ef

Fig. 1 Identifcation of aberrantly methylation-regulated genes from GEO and TCGA databases. Aberrant DNA methylation between CRC tumors and adjacent normal tissues in GSE17648 (a) and GSE29490 (b); c Ectopically expressed genes in CRC tumor tissues from TCGA database; d Venn diagram of DNA methylation and expression in tumor tissue; e, f Heat map showing the diferential expression and methylation of 14 genes between tumors and adjacent normal tissues from TCGA database Zhang et al. J Transl Med (2021) 19:98 Page 5 of 13 < 0.001 < 0.001 < 0.001 < 0.001 0.004 < 0.001 < 0.001 < 0.001 < 0.001 < 0.001 < 0.001 < 0.001 0.708 < 0.001 P − 0.431 − 0.222 − 0.492 − 0.431 − 0.357 − 0.404 − 0.455 − 0.411 − 0.383 − 0.351 − 0.594

0.246

0.138

0.018

Cor Correlation < 0.001 < 0.001 < 0.001 < 0.001 < 0.001 < 0.001 < 0.001 < 0.001 < 0.001 < 0.001 < 0.001 < 0.001 < 0.001 < 0.001 P ± 1.993 ± 2.171 ± 1.826 ± 0.127 ± 0.874 ± 1.291 ± 1.885 ± 1.284 ± 1.805 ± 1.259 ± 1.367 ± 1.555 ± 2.459 ± 1.007 4.669 3.451 2.442 0.437 0.667 1.260 5.791 1.920 2.982 1.090 1.096 3.660 3.036 0.753 Tumor ± 1.425 ± 1.064 ± 1.170 ± 0.016 ± 0.969 ± 0.848 ± 0.826 ± 0.590 ± 0.906 ± 1.039 ± 1.915 ± 0.506 ± 1.658 ± 0.981 8.333 9.030 6.537 0.232 3.120 5.336 9.050 6.019 7.715 4.470 3.389 7.536 7.837 4.678 Normal Expression < 0.001 < 0.001 < 0.001 < 0.001 0.016 < 0.001 < 0.001 < 0.001 < 0.001 < 0.001 0.131 < 0.001 < 0.001 < 0.001 P ± 0.155 ± 0.093 ± 0.059 ± 0.118 ± 0.081 ± 0.089 ± 0.079 ± 0.116 ± 0.075 ± 0.075 ± 0.104 ± 0.053 ± 0.053 ± 0.091 0.340 0.390 0.359 0.359 0.534 0.517 0.571 0.461 0.450 0.451 0.557 0.363 0.325 0.443 Tumor ± 0.009 ± 0.015 ± 0.010 ± 0.130 ± 0.026 ± 0.022 ± 0.017 ± 0.137 ± 0.034 ± 0.038 ± 0.027 ± 0.022 ± 0.150 ± 0.031 0.121 0.301 0.431 0.175 0.577 0.324 0.393 0.242 0.283 0.270 0.316 0.259 0.308 0.156 Normal Methylation Correlation between gene expression and methylation of CRC in TCGA database in TCGA of CRC and methylation expression between gene Correlation EPHA7 PRIMA1 ADCYAP1R1 NOVA1 LRRC3B TMEFF2 GFRA1 RXRG RSPO2 PHOX2A KCNA1 HPSE2 GPM6A KHDRBS2 1 Table Gene Zhang et al. J Transl Med (2021) 19:98 Page 6 of 13

Results exp) groups. As shown in Fig. 2b and Additional fle 2: Fig Screening of methylation‑regulated genes from the GEO S1J–R, only HPSE2 could be used to clearly distinguish the and TCGA databases survival of patients. Patients with the hypermethylation– We selected 2 methylation datasets (GSE17648 and low expression of HPSE2 had signifcantly poor survival GSE29490) to screen for aberrantly methylation-regulated (P = 0.032). genes. In accordance with the criteria, 692 diferentially methylated genes (136 hypermethylated and 557 hypo- Hypermethylation‑low expression of HPSE2 methylated genes) between tumors and normal tissues was an independent predictor of the poor outcome from the GSE17648 dataset (Fig. 1a and Additional fle 1: of patients with CRC​ Table S1) and 514 diferentially methylated genes (176 HPSE2 was signifcantly abnormally methylated hypermethylated and 338 hypomethylated genes) from the (GSE17648 and GSE29490) and expressed in tumor tissue GSE29490 dataset were identifed (Fig. 1b). Additionally, (TCGA database) as shown in Fig. 3a–d. Ten, patients 1026 aberrantly expressed genes (663 upregulated and 363 with CRC were divided into the hypermethylation-low downregulated genes) were obtained from TCGA data- expression and hypomethylation-high expression groups base (Fig. 1c and Additional fle 1: Table S1). Finally, sixteen in accordance with the median of HPSE2 (the expression hypermethylated–lowly expressed and 0 hypomethylated– level was 3.660 and the methylation level was 0.367). Fur- highly expressed genes were screened out (Fig. 1d) by tak- ther stratifcation by TNM stage revealed that patients ing the intersection. with HPES2 hypermethylation had signifcantly short- We mapped the methylation and expression data of 16 ened survival in stage III/IV (P < 0.05) but not in stages I/ genes from TCGA database, which contained 407 CRC tis- II (P > 0.05; Fig. 3e, f). Univariate Cox regression revealed sues and 21 adjacent tissues (Fig. 1e, f). NPYR and CHR2 that HPES2 hypermethylation–low expression was asso- were deleted due to the absence of corresponding methyla- ciated with an increased risk of cancer-related death (rel- tion information. ative risk [RR]:1.412; 95% CI 1.043–1.913, P = 0.026). As Te Pearson correlation between expression and meth- expected, T-N-M stage was also a signifcant prognostic ylation was further evaluated, and 10 out of the 14 methyla- factor. In particular, multivariate Cox regression analy- tion-regulated genes were screened in accordance with the sis revealed that HPSE2 methylation–expression was an criteria (cor < − 0.3, Table 1, Fig. 2a and Additional fle 2: independent risk factor for shortened survival among Fig. S1A-I). We considered that those genes might partici- patients with CRC (RR: 1.494; 95% CI 1.056 to 2.114, pate in the development of tumors and afect the progno- P = 0.024) (Fig. 3g). Tese fndings indicated that the sis of patients. Kaplan–Meier plot analysis was therefore hypermethylation–expression of HPSE2 was predictive performed and 403 patients with CRC were divided into of the poor prognosis of patients with CRC, especially hypomethylation–high expression (hypo and high exp) those in the late stages of the disease. and hypermethylation–low expression (hyper and low

ab

Fig. 2 Expression and methylation of HPSE2 was associated with 5-year overall survival in CRC patients. a Pearson correlation between HPSE2 expression and methylation; b Patients with hypermethylation–low expression of HPSE2 have a worse prognosis Zhang et al. J Transl Med (2021) 19:98 Page 7 of 13

CpG islands methylation of HPSE2 in CRC​ showed a signifcant correlation with HPSE2 expression We further evaluated CpG islands methylation status (cor < − 0.3; P < 0.05) (Fig. 4b) and were signifcantly dif- within the 3 kb region of the HPSE2 promoter on the ferentially expressed between cancer and adjacent nor- basis of TCGA database. Among the 49 obtained meth- mal tissues (Fig. 4c). We further verifed these sites in ylation sites (Fig. 4a and Additional fle 3: Table S2), 13

ab c *** .5 *** Normal Normal *** Normal

0. 5 Tumor Tumor Tumor n 0. 40 .4 .3 0. 30 0. 20 .2 .1 HPSE2 methylation HPSE2 methylation 036 9 HPSE2 expression 0. 10 0. 00 GSE17648 GSE29490 TCGA de f *** Normal TNM stage1/2 TNM stage3/4

.5 Tumor 1. 0 1. 0 0. 40 .5 .3 0. 5

Survival rate P = 0.067PSurvival rate = 0.006 HPSE2 methylation

0. 20 HPSE2 hypo & high exp HPSE2 hypo & high exp HPSE2 hyper & low exp HPSE2 hyper & low exp 0 00 TCGA 0510 15 0510 15 Time (years) Time (years) g

Univariate Multivariate Study Hazard Ratio(95 %CI) P Value Hazard Ratio(95 %CI) P Value

Age 1.810(0.898−3.651) 0.097

Gender 1.351(0.738−2.473) 0.329

T 2.720(1.472−5.028) 0.001 2.760(1.348−5.650) 0.005

N 2.012(1.109−3.653) 0.021 1.646(0.780−3.472) 0.191

M 3.056(1.542−6.058) 0.001 2.142(1.004−4.571) 0.049

HPSE2 1.412(1.043−1.913) 0.026 1.494(1.056−2.114) 0.024 methy & exp

0.511.522.533.5 44.5 55.5 6 11.5 22.5 33.5 44.5 55.5 6 Hzard Ratio(HR) Hzard Ratio(HR)

Fig. 3 HPSE2, which was regulated by promoter DNA methylation, could be an independent prognostic factor. a Methylation levels of HPSE2 in 22 cases of CRC tissues in GSE17648; b Methylation levels of HPSE2 in 26 cases of matched CRC tumors and adjacent normal tissues in GSE29490; HPSE2 expression (c) and methylation (d) in TCGA database; e, f Kaplan–Meier curves showing that hypermethylation–low expression of HPSE2 was associated with the shortened survival of CRC patients at the late stages (f) but not at the early stages (e); g Univariate and multivariate COX regression model revealed that HPSE2 could be an independent prognostic risk factor. ***P < 0.001 Zhang et al. J Transl Med (2021) 19:98 Page 8 of 13

GSE48484 and found that CpG islands methylation level Tis phenomenon were verifed in an in vitro experi- were higher in cancer and adenoma tissues (Fig. 4d). ment. As shown in Fig. 5c, after 5 days of demethyla- As shown in Fig. 5a, the diference was further observed tion treatment, HPSE2 CpG island methylation began to in 6 pairs of samples in GSE77965. After treatment with decrease continuously in HCT116 from the GSE51810 5-aza-2′-deoxycitidine (DAC), CpG islands methylation dataset. decreased in peripheral blood (Fig. 5b). y y y y y y y y y y y y y y y y y y y y y y y y y y y y y y y y y y a y y y y y y y y y y y y y y y d Sample HPSE 2 cg20552296|meth cg19043522|meth cg06408053|meth cg00877329|meth cg13343639|meth cg25691326|meth cg24134845|meth cg10441934|meth cg10580473|meth cg00612625|meth cg11400953|meth cg18754857|meth cg09511126|meth cg17508300|meth cg26329178|meth cg25034395|meth cg26067760|meth cg19316579|meth cg02800607|meth cg19622451|meth cg07198258|meth cg15610020|meth cg25437411|meth cg24926711|meth cg18499443|meth cg13737332|meth cg13690864|meth cg08529260|meth cg16825290|meth cg08927006|meth cg17868515|meth cg07717704|meth cg22823644|meth cg03388193|meth cg24591300|meth cg25651562|meth cg18577511|meth cg17863743|meth cg16745930|meth cg10943524|meth cg22892437|meth cg27247731|meth cg04070995|meth cg16298041|meth cg03236397|meth cg14270857|meth cg19428722|meth cg16401360|meth cg14251734|meth

HPSE2 1 cg20552296|methy cg19043522|methy cg06408053|methy cg24926711 Normal cg00877329|methy ***

cg13343639|methy 0.8 *** cg25691326|methy cg24134845|methy cg10441934|methy cg10580473|methy cg00612625|methy 0.6 Adenoma cg11400953|methy cg18754857|methy cg09511126|methy cg17508300|methy cg26329178|methy 0.4 cg25034395|methy cg18499443 Tumor

cg26067760|methy *** cg19316579|methy cg02800607|methy *** cg19622451|methy 0.2 cg07198258|methy cg15610020|methy cg25437411|methy cg24926711|methy cg18499443|methy 0 cg13737332|methy cg13690864|methy cg08529260|methy cg16825290|methy cg17863743 cg08927006|methy −0.2 ***

cg17868515|methy *** cg07717704|methy cg22823644|methy cg03388193|methy cg24591300|methy −0.4 cg25651562|methy cg18577511|methy cg17863743|methy cg16745930|methy cg10943524|methy −0.6 cg22892437|methy cg27247731|methy cg17508300 cg04070995|methy ***

cg16298041|methy *** cg03236397|methy −0.8 cg14270857|methy cg19428722|methy cg16401360|methy cg14251734|methy −1

cg16825290 ***

b *** cg16401360 1.0 0.59 cg14270857 0.5 cg03236397 0.70 0.56 0.0 cg16401360 ***

cg17863743 0.61 0.64 0.91 −0.5 *** −1.0 cg03388193 0.57 0.59 0.74 0.56 cg08927006 0.52 0.86 0.65 0.64 0.74 cg14270857 0.58 0.55 0.71 0.53 0.61 0.71 ***

cg16825290 ** * cg13690864 0.88 0.49 0.57 0.63 0.46 0.58 0.64 cg13737332 0.68 0.71 0.72 0.58 0.90 0.55 0.57 0.86

0.73 0.46 0.52 0.90 0.52 0.80 0.60 0.57 0.71 cg13737332 cg18499443 B ** * ** * cg24926711 0.66 0.69 0.60 0.63 0.65 0.52 0.66 0.56 0.57 0.62 cg17508300 0.53 0.56 0.66 0.63 0.63 0.58 0.57 0.64 0.60 0.57 0.59 cg09511126 0.58 0.64 0.72 0.82 0.64 0.73 0.77 0.58 0.90 0.62 0.65 0.93 cg13690864 ** * ** * HPSE2 −0.39 −0.33 −0.35 −0.33 −0.37 −0.33 −0.38 −0.34 −0.31 −0.39 −0.32 −0.32 −0.39

cg09511126

c ** *

Tpye Tpye ** * cg03388193 Normal cg24926711 Tumor cg13737332 cg08927006 ** *

cg17863743 0.8 ** * cg18499443 cg08927006 0.6 cg09511126 cg03388193

0.4 ** * cg16401360 ** *

cg03236397 0.2 cg14270857 cg17508300 cg03236397 ** * cg13690864 ** * cg16825290 0.00 0.25 0.50 0.75 Methylation Fig. 4 CpG islands methylation of the HPSE2 promoter. a Heat map of 49 methylated CpG islands located in the promoter region of HPSE2 from TCGA database; b Pearson correlation of methylation sites and HPSE2 expression with cor < 0.3 were selected; c Diferentially methylated − CpG islands between normal and tumor tissues from TCGA database; d Diferentially methylated CpG islands of the HPSE2 promoter among adenocarcinoma, adenoma, and normal tissues from the GSE68484 dataset. ***P < 0.001 Zhang et al. J Transl Med (2021) 19:98 Page 9 of 13

HPSE2 was expressed at signifcantly low levels in tumor Ectopic expression of HPSE2 suppressed CRC cell growth, tissues migration, and cell cycle progression We detected HPSE2 expression in CRC samples and Te expression of HPSE2 was detected in 5 CRC cell peripheral blood. As shown in Fig. 6a, HPSE2 mRNA lines and 1 normal intestinal epithelial cell line. As shown was downregulated in tumor tissues. However, HPSE2 in Fig. 7a, among all detected cells, HPSE2 was highly expression in the peripheral blood of patients with CRC expressed in NCM460 and relatively lowly expressed in did not signifcantly difer from that of healthy controls HCT116 and SW480. Ten, the HPSE2-overexpression (Fig. 6b). HPSE2 protein levels were also detected in 10 vector was stably transfected into SW480 and HCT116 pairs of tumor tissues (Fig. 6c). cells with an empty vector as a control (Fig. 7b). Te ectopic expression of HPSE2 signifcantly inhibited

ab

c

Fig. 5 Changes in CpG island methylation in the HPSE2 promoter after DCA treatment. a CpG island methylation in 6 matched cancers and adjacent tissues; b Changes in methylation level in peripheral blood of mice treated with DCA in GSE77965 dataset; c, d Changes in methylation levels after adding DAC to HCT116 cells from the GSE51810 dataset Zhang et al. J Transl Med (2021) 19:98 Page 10 of 13

cell growth as evidenced by the CCK8 assay results for HPSE2 could regulate the p53/p21 signaling pathway HCT116 (Fig. 7c) and SW480 (Fig. 7d). HPSE2 overex- We performed GSEA enrichment analysis to fur- pression markedly suppressed the migration capability of ther explore the signal pathways that HPSE2 might be HCT116 and SW480 cells (Fig. 7e, f). Cell cycle results involved in. Among all tumor-related enrichment path- showed that HPSE2 overexpression could block the cell ways, HPSE2 could regulate the p53 pathway (p < 0.001) cycle mainly in the G1 phase (Fig. 7g). and the cell cycle (p < 0.001) (Fig. 7l and Additional fle 4: We built subcutaneous xenograft tumor models by sub- Table S3). Terefore, we performed Western blot analysis cutaneously injecting HPSE2-transfected and empty-vec- and confrmed that HPSE2 overexpression could upregu- tor-transfected HCT116 cells into nude mice to examine late p53/p21 levels in HCT116 and SW480 cells (Fig. 7m). the efect of HPSE2 on CRC growth in vivo. Tumor vol- ume was monitored and compared between the 2 groups. Discussion As shown in Fig. 7h–j, tumor volume was signifcantly As one of the common forms of molecular alterations in suppressed after HPSE2 overexpression. Te immunohis- carcinogenesis [15], DNA methylation is involved in the tochemical staining results for KI67 revealed signifcantly occurrence and development of tumors, including CRC reduced cell proliferation after HPSE2 overexpression [16–19]. CRC is characterized by promoter hypermeth- (Fig. 7k). Te above results indicated that HPSE2 might ylation and the CIMP phenotype of tumor suppressor be a potential tumor suppressor gene in CRC. genes [6]. Tumor-related aberrant DNA methylation in the serum of patients with cancer can be used as a molecular marker for the survival and recurrence of CRCs [20–23]. Hence, the identifcation of novel methylation-regulated

ab

c

Fig. 6 HPSE2 was signifcantly downregulated in cancer tissues. We detected HPSE2 expression in CRC samples and peripheral blood. HPSE2 was aberrantly expressed in CRC tissues (a) but not in peripheral blood (b); HPSE2 protein showed diferential expression in 10 pairs of CRC tumors and normal tissues (c) Zhang et al. J Transl Med (2021) 19:98 Page 11 of 13

d abSW480 HCT116 c SW480 HCT116 800 20 5 12 ** 1400 Vector ** Vector *** 700 *** l l l 1200 10 *** 4 LV-HPSE2 LV-HPSE2 600 *** *** 15 1000 500 8

800 3 400 10 6 2.0 2.0 *** *** 2 ** *** 1.5 1.5 4 *** 5 1.0 1.0 Relative growth rate Relative growth rate 1 Relative HPSE2 leve Relative HPSE2 leve Relative HPSE2 leve * ** 2 NS 0.5 0.5

0 0.0 0.0 0 0 Vector LV-HPSE2 Vector LV-HPSE2 01234 01234 116 LOVO DLD1 Days SW480 SW620 Days HCT MNC460

ef g 80 ** SW480HCT116 Vector 60 LV-HPSE2 60 40 *** *** 40 *** ctor ctor 30 SW48 0 SW48 0 20 Ve Ve 40 0 G1 S G2 80 20 * Vector 60 LV-HPSE2 20 16 16

-HPSE2 40 -HPSE2 10 ** LV HCT1 HCT1 LV 20

0 0 0 G1 SG2 Vector LV-HPSE2VectorLV-HPSE2VectorLV-HPSE2

hiVector j k KI67 )

3 1.8 Vector 1.5 LV-HPSE2 * Vector 1.2 0.9 ctor 0.6

* Ve 0.3 mor volume (m m LV-HPSE2 0.0 Tu 012345 LV-HPSE2 Time(Weeks) 24 Vector

) 22 LV-HPSE2

20 -HPSE2 18 LV 16

Mice weight (g 14 012345 Time(Weeks) lm SW480HCT116 .6

p53

0. 40 Cell cycle DNA replication

.2 Natural killer cell mediated cytotoxocity p53 ichment Scor e r P53 signaling pathway p21 En

0. 00 Pyrimidine metabolism

GAPDH

High risk<−−−−−−−−−−−>Low risk Vector LV-HPSE2 Vector LV-HPSE2 Fig. 7 HPSE2 performs a tumor suppressive function by activating the p53/ p21 signaling cascade. a HPSE2 mRNA expression in 5 CRC cell lines and 1 intestinal epithelial cell; b HPSE2 overexpression in SW480 and HCT116 was confrmed through qRT-PCR; c, d HPSE2 overexpression signifcantly inhibited SW480 (c) and HCT116 (d) proliferation; e, f HPSE2 overexpression inhibited SW480 (e) and HCT116 (f) migration; g HPSE2 blocked the SW480 and HCT116 cell cycle in the G1–S stage; h, i Images of tumor specimens; j HPSE2 overexpression signifcantly suppressed tumorigenicity in vivo; k Immunohistochemistry results for mouse tumor tissues: ki67 expression was decreased in the HPSE2-overexperssing and control group; l GSEA plot showed pathways that were signifcantly related to HPSE2 (p < 0.05); m Western blot analysis showed that HPSE2 overexpression could upregulate p53/p21 levels in HCT116 and SW480. *P < 0.05, **P < 0.01, ***P < 0.001 Zhang et al. J Transl Med (2021) 19:98 Page 12 of 13

oncogenes and tumor suppressor in CRC may provide Conclusion insights into epigenetic mechanisms and help identify In summary, we identifed HPSE2, a novel tumor sup- new therapeutic targets. pressor gene that is frequently inactivated through pro- According to previous analysis in the GEO database, moter methylation in CRC. HPSE2 performs a tumor TCN1 and TGFBI were identifed as hub genes regu- suppressive function by activating the p53/ p21 signaling lated by DNA methylation. Teir functions have been cascade. Te promoter hypermethylation of HPSE2 is a confrmed in CRC which further indicates the reliability potential therapeutic target in patients with CRC, espe- of our analysis. To fnd new key genes, we comprehen- cially those with late-stage CRC. sively analyzed multiple datasets and found that the sig- nifcantly low expression of HPSE2 in CRC tissues was Supplementary Information regulated by promoter methylation. Aberrant methyla- The online version contains supplementary material available at https​://doi. tion-regulated HPSE2 was correlated with patient prog- org/10.1186/s1296​7-021-02770​-0. nosis and was more pronounced in patients with CRC in stage III/IV. Multivariate COX regression analysis Additional fle 1: Table S1. Screening of methylation-regulated genes from the GEO and TCGA databases. showed that methylation-regulated HPSE2 could be used Additional fle 2: Fig S1. Screening for methylation-regulated genes as an independent prognostic risk factor. related to prognosis. A–I: Pearson correlation between gene expression HPSE2 encodes heparanase, an that degrades and methylation. Ten genes with a correlation greater than 0.3 are shown; heparin sulfate proteoglycans, and is located on the J-R: Kaplan–Meier plot analysis of the relationship between methylation- regulated genes and patient prognosis. extracellular matrix and cell surface [24]. Mutations in Additional fle 3: Table S2. Correlation between HPSE2 expression and HPSE2 may be related to urofacial syndromes [25–27]. CpG island methylation of CRC in TCGA database. Tis protein may function in angiogenesis and tumor Additional fle 4: Table S3. GSEA enrichment analysis. progression by participating in biological processes, Additional fle 5. Codes for screening of diferential expressed genes and such as the remodeling of the extracellular matrix [28, methylation data. 29]. It may act as a suppressor gene in tumors, includ- ing breast cancer [30, 31] and cervical cancer [32]. Acknowledgements However, the expression and role of HPSE2 in CRC has Not applicable. not yet been reported. We investigated the function of HPSE2 in CRC in vitro Authors’ contributions JF and JW conceived and designed the experiments. HZ and CX analysed the and in vivo. Cell proliferation capability was signifcantly data. CS and JZ conducted in vitro experiments. TQ, RM and ZW reviewed suppressed in HCT116 and SW480 cells with the ectopic the draft. FJ revised the manuscript. All authors read and approved the fnal overexpression of HPSE2 compared with that in cells manuscript. transfected with the empty vector. Consistently, com- Funding pared with the control group, the xenografts with HPSE2 Jiangsu Provincial Key Research Development Program, Grant/Award Num- overexpression showed decreased tumor volumes. In bers: BE2016794 and BE2016795. addition, HPSE2 could inhibit tumor cell migration, sug- Availability of data and materials gesting that HPSE2 might afect CRC metastasis. Cell The data sets used and/or analyzed during the current study are available cycle experiments confrmed that HPSE2 could cause from the GEO database (http://www.ncbi.nlm.nih.gov/ geo/) and TCGA data- base (https​://cance​rgeno​me.nih.gov/). The methylation data used in the study cell cycle arrest mainly in the G1 phase. Te above results (GSE17648 and GSE29490) are available in a public repository from NCBI (https​ ://www.ncbi.nlm.nih.gov/geo/query​/acc.cgi?acc GSE17​648; https​://www. indicated that abnormally expressed HPSE2 played a = tumor suppressive role in CRC processes. ncbi.nlm.nih.gov/geo/query​/acc.cgi?acc GSE29​490). The CpG island methyla- tion data (GSE68484 and GSE51810) are available= in a public repository from We performed GSEA enrichment analysis to further NCBI (https​://www.ncbi.nlm.nih.gov/geo/query​/acc.cgi?acc GSE68​484; https​ ://www.ncbi.nlm.nih.gov/geo/query​/acc.cgi?acc GSE51​810=). All relevant data explore the potential mechanisms of HPSE2 and found = that HPSE2 could afect the cell cycle and p53 signal- are available from the authors (Additional fle 5). ing pathway. P53 and its downstream target p21 play an Declarations important role in suppressing G1–S cell cycle transition [33, 34]. Tus, we performed Western blot analysis and Ethics approval and consent to participate Not applicable. confrmed that HPSE2 overexpression could activate the p53 / p21 signaling pathway. Consent for publication Not applicable.

Competing interests The authors declare that they have no competing interests. Zhang et al. J Transl Med (2021) 19:98 Page 13 of 13

Author details 16. Jin P, Song Y, Yu G. The Role of Abnormal Methylation of Wnt5a gene 1 Department of General Surgery, Jiangsu Cancer Hospital, Jiangsu Institute promoter regions in human epithelial ovarian cancer: a clinical and of Cancer Research, The Afliated Cancer Hospital of Nanjing Medical Univer- experimental Study. Anal Cell Pathol (Amst). 2018;2018:6567081. sity, Nanjing 210000, Jiangsu, People’s Republic of China. 2 Research Center 17. Nephew KP, Huang TH-M. Epigenetic gene silencing in cancer initiation for Clinical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer and progression. Cancer Lett. 2003;190(2):125–33. Research, The Afliated Cancer Hospital of Nanjing Medical University, 42 18. Towle R, Truong D, Hogg K, et al. Global analysis of DNA methylation Baiziting, Nanjing 210000, Jiangsu, People’s Republic of China. 3 Department changes during progression of oral cancer. Oral Oncol. 2013;49:1033–42. of Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 19. Farkas SA, Milutin-Gašperov N, Grce M, Nilsson TK. Genome-wide DNA The Afliated Cancer Hospital of Nanjing Medical University, Nanjing 210000, methylation assay reveals novel candidate biomarker genes in cervical Jiangsu, People’s Republic of China. 4 Jiangsu Key Laboratory of Molecular cancer. Epigenetics. 2013;8:1213–25. and Translational Cancer Research, Jiangsu Cancer Hospital, Jiangsu Institute 20. Hibi K, Goto T, Shirahata A, et al. Detection of TFPI2 methylation in the of Cancer Research, The Afliated Cancer Hospital of Nanjing Medical Univer- serum of colorectal cancer patients. Cancer Lett. 2011;311(1):96–100. sity, 42 Baiziting, Nanjing 210000, Jiangsu, People’s Republic of China. 21. Herbst A, Wallner M, Rahmig K, et al. Methylation of helicase-like transcription factor in serum of patients with colorectal cancer is an Received: 8 November 2020 Accepted: 25 February 2021 independent predictor of disease recurrence. Eur J Gastroenterol Hepatol. 2009;21(5):565–9. 22. Lecomte T, Berger A, Zinzindohoué F, et al. Detection of free-circulating tumor-associated DNA in plasma of colorectal cancer patients and its association with prognosis. Int J Cancer. 2002;100(5):542–8. References 23. Nakayama H, Hibi K, Takase T, et al. Molecular detection of p16 promoter 1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global methylation in the serum of recurrent colorectal cancer patients. Int J Cancer Statistics 2018: GLOBOCAN estimates of incidence and Cancer. 2003;105(5):491–3. mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 24. Oskarsson T. Extracellular matrix components in breast cancer progres- 2018;68(6):394–424. sion and metastasis. Breast. 2013;22(Suppl 2):S66-72. 2. Pochapin MB. 2017 Emily Couric memorial lecture: Colorectal Cancer: Pol- 25. Daly SB, Urquhart JE, Hilton E, et al. Mutations in HPSE2 cause urofacial yps, Prevention, and Progress. Am J Gastroenterol. 2018;113(12):1772–7. syndrome. Am J Hum Genet. 2010;86(6):963–9. 3. Kim MS, Lee J, Sidransky D. DNA methylation markers in colorectal cancer. 26. Pang J, Zhang S, Yang P, et al. Loss-of-function mutations in HPSE2 Cancer Metastasis Rev. 2010;29(1):181–206. cause the autosomal recessive urofacial syndrome. Am J Hum Genet. 4. Puccini A, Berger MD, Naseem M, et al. Colorectal cancer: epigenetic 2010;86(6):957–62. alterations and their clinical implications. Biochim Biophys Acta Rev 27. Mahmood S, Beetz C, Tahir MM, et al. First HPSE2 missense mutation in Cancer. 2017;1868(2):439–48. urofacial syndrome. Clin Genet. 2012;81(1):88–92. 5. McMahon KW, Enusha K, Nita A. The Roles of DNA Methylation in the 28. Poltavets V, Kochetkova M, Pitson SM, Samuel MS. The role of the extracel- Stages of Cancer. Cancer J. 2017;23(5):257–61. lular matrix and its molecular and cellular regulators in cancer cell plastic- 6. Wong CC, Li W, Chan B, Yu J. Epigenomic biomarkers for prognostica- ity. Front Oncol. 2018;8:431. tion and diagnosis of gastrointestinal cancers. Semin Cancer Biol. 29. Zcharia E, Metzger S, Chajek-Shaul T, et al. Molecular properties and 2019;55:90–105. involvement of heparanase in cancer progression and mammary gland 7. Janson W, Tse T, Jenkins J, Chionh F, Mariadason JM. Aberrant DNA morphogenesis. J Mammary Gland Biol Neoplasia. 2001;6(3):311–22. methylation in colorectal cancer: what should we target? Trends Cancer. 30. Wu B, Liu G, Jin Y, et al. miR-15b-5p Promotes Growth and Metastasis in 2017;3(10):698–712. Breast Cancer by Targeting HPSE2. Front Oncol. 2020;10:108. 8. Sakai E, Nakajima A, Kaneda A. Accumulation of aberrant DNA methyla- 31. Fu J, Khaybullin R, Zhang Y, Xia A, Qi X. Gene expression profling leads to tion during colorectal cancer development. World J Gastroenterol. discovery of correlation of matrix metalloproteinase 11 and heparanase 2 2014;20(4):978–87. in breast cancer progression. BMC Cancer. 2015;15:473. 9. Draht MXG, Goudkade D, Koch A, et al. Prognostic DNA methylation 32. Marques RM, Focchi GR, Theodoro TR, et al. The immunoexpression of markers for sporadic colorectal cancer: a systematic review. Clinical heparanase 2 in normal epithelium, intraepithelial, and invasive squa- Epigenetics. 2018;10:35. mous neoplasia of the cervix. J Low Genit Tract Dis. 2012;16(3):256–62. 10. Cancer Genome Atlas Research Network. Comprehensive molecu- 33. Wang K, Liang Q, Li X, et al. MDGA2 is a novel tumour suppressor lar characterization of human colon and rectal cancer. Nature. cooperating with DMAP1 in gastric cancer and is associated with disease 2012;487(7407):330–7. outcome. Gut. 2016;65(10):1619–31. 11. Cancer Genome Atlas Research Network. Comprehensive molecular char- 34. Kachnic LA, Wu B, Wunsch H, et al. The ability of p53 to activate down- acterization of gastric adenocarcinoma. Nature. 2014;513(7517):202–9. stream genes p21(WAF1/cip1) and MDM2, and cell cycle arrest following 12. Zhang H, Dong S, Feng J. Epigenetic profling and mRNA expression DNA damage is delayed and attenuated in scid cells defcient in the reveal candidate genes as biomarkers for colorectal cancer. J Cell Bio- DNA-dependent protein kinase. J Biol Chem. 1999;274(19):13111–7. chem. 2019;120(6):10767–76. 13. Yu JM, Sun W, Wang ZH, et al. XTRIB3 supports breast cancer stemness by suppressing FOXO1 degradation and enhancing SOX2 transcription. Nat Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in pub- Commun. 2019;10(1):5720. lished maps and institutional afliations. 14. Subramanian A, Kuehn H, Gould J, Tamayo P, Mesirov JP. GSEA-P: a desktop application for Gene Set Enrichment Analysis. Bioinformatics. 2007;2323(23):3251–3. 15. Baylin SB, Belinsky SA, Herman JG. Aberrant methylation of gene promot- ers in cancer—concepts, misconcepts, and promise. J Natl Cancer Inst. 2000;92(18):1460–1.