Open Access

Association between the expression levels of ADAMTS16 and BMP2 and tumor budding in hepatocellular carcinoma

  • Authors:
    • Di Jiang
    • Shaoshao Xu
    • Chuanpeng Zhang
    • Chuanbing Hu
    • Lei Li
    • Mingming Zhang
    • Haiyan Wu
    • Dongchang Yang
    • Yanrong Liu
  • View Affiliations

  • Published online on: April 27, 2023     https://doi.org/10.3892/ol.2023.13842
  • Article Number: 256
  • Copyright: © Jiang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tumor budding (TB) has become a crucial factor for predicting the malignancy grade and prognostic outcome for multiple types of solid cancer. Studies have investigated the prognostic value of TB in hepatocellular carcinoma (HCC). However, its molecular mechanism in HCC remains unclear. To the best of our knowledge, the present study was the first to compare the expression of differentially expressed genes (DEGs) between TB‑positive (TB‑pos) and TB‑negative HCC tissues. In the present study, total RNA was extracted from 40 HCC tissue specimens and then sequenced. According to Gene Ontology (GO) functional annotation, upregulated DEGs were markedly associated with embryonic kidney development‑related GO terms, which suggested that the TB process may at least partly mimic the process of embryonic kidney development. Subsequently, two genes, a disintegrin and metalloproteinase domain with thrombospondin motifs 16 (ADAMTS16) and bone morphogenetic protein 2 (BMP2), were screened and verified through immunohistochemical analysis of HCC tissue microarrays. According to the immunohistochemical results, ADAMTS16 and BMP2 were upregulated in TB‑pos HCC samples, and BMP2 expression was increased in budding cells compared with the tumor center. Additionally, through cell culture experiments, it was demonstrated that ADAMTS16 and BMP2 may promote TB of liver cancer, thus promoting the malignant progression of liver cancer. Further analysis revealed that ADAMTS16 expression was associated with necrosis and cholestasis, and BMP2 expression was associated with the Barcelona Clinic Liver Cancer stage and the vessels encapsulating tumor clusters. Overall, the findings of the present study provided insights into the possible mechanisms of TB in HCC and revealed potential anti‑HCC therapeutic targets.

Introduction

According to the 2020 Global Cancer Statistics, liver cancer ranks 6th in terms of global morbidity and 3rd among causes of cancer-associated mortality worldwide (1). Hepatocellular carcinoma (HCC) is the most frequent subtype of primary liver cancer that occurs in 75–85% of patients with liver cancer (2). Despite the great progress made in anti-HCC therapy, HCC is associated with a poor prognostic outcome (3). Tumor metastasis is an important factor contributing to the poor HCC prognosis (4). Tumor budding (TB) is defined as dissociation of isolated cancer cells and/or discrete clusters (<5 cancer cells), and its prognostic role was first reported in colorectal cancer (CRC) (5). In addition to CRC, TB has been observed in various cancer types and predicts a poor prognosis, including in esophageal (6), nasopharyngeal (7), lung (8) and pancreatic cancer (9). Furthermore, TB has been reported to be associated with epithelial-mesenchymal transition (EMT), which is a critical event that supports tumor migration and invasion (10,11). However, the impact of TB on HCC remains poorly understood. Only two studies have analyzed the prognostic value of TB in HCC (12,13), while the mechanistic basis of TB in HCC remains unclear.

A disintegrin and metalloproteinase domain with thrombospondin motifs 16 (ADAMTS16) belongs to the ADAMTS family. Its role has been investigated in esophageal squamous cell carcinoma (14) and CRC (15,16). However, to the best of our knowledge, the role of ADAMTS16 in HCC has not yet been studied. Bone morphogenetic protein 2 (BMP2), which belongs to the transforming growth factor-β superfamily, participates in different cancer occurrence and development processes (1719). Certain studies have indicated that BMP2 enhances HCC cell growth, invasion and migration (20,21). However, the relationship between BMP2 and TB in HCC remains to be studied.

The aim of this study was to identify differentially expressed genes (DEGs) in TB-positive (TB-pos) HCC tissue samples relative to TB-negative (TB-neg) HCC tissue samples through bioinformatics analysis, and then explore the potential mechanism of TB in HCC. In the present study, it was demonstrated that ADAMTS16 and BMP2 levels were significantly increased in the TB-pos HCC samples, and BMP2 expression was significantly increased in budding cells compared with the tumor center. Additionally, it was demonstrated that overexpression (OE) of ADAMTS16 and BMP2 promoted invasion of HepG2 cells, which implied that ADAMTS16 and BMP2 may regulate TB in liver cancer. The association of ADAMTS16 and BMP2 expression with clinicopathological characteristics and patient survival time were also analyzed. The findings of the present study provide novel mechanistic insights into the role of ADAMTS16 and BMP2 in HCC.

Materials and methods

Patients and tissues

Paraffin-embedded tumor specimens from patients with HCC (n=308) were retrospectively collected from The Affiliated Hospital of Jining Medical University (Jining, China) between June 2013 and August 2019. The clinical and pathological details of the patients were retrospectively reviewed by accessing the hospital's existing electronic medical record system. Ultimately, 266/308 (86%) of patients with HCC with complete clinical information were selected for the subsequent clinical significance analysis. In addition, of the 308 patients, 66 were excluded from the survival analysis due to a lack of follow-up. And additional 40 HCC tissue samples were prospectively collected from The Affiliated Hospital of Jining Medical University between February 2020 and June 2021 for transcriptome sequencing. All samples were histopathologically and clinically confirmed as HCC tissues. The histology, diagnostic methods, and α-fetoprotein (AFP) levels were reviewed for each case of HCC identified to eliminate cases of metastatic cancer or other primary liver cancer types. Microscopically, the HCC cells were arranged in solid nests, trabeculae, acinar or pseudoglandular structures. Papillary structures were occasionally seen. Abundant sinusoidal capillaries could be seen between the tumor alveolus, with steatosis and eosinophilic bodies. Ethics approval was provided by The Ethics Committee of The Affiliated Hospital of Jining Medical University (Jining, China; ethical approval no. 2021C145). Each participant (>18 years of age) in the study provided written informed consent and signed informed consent forms.

Cell culture and lentiviral infection

The human hepatoblastoma cell line, HepG2, was purchased from ATCC via the Shanghai Huiying Biological Technology Co., Ltd. The cell line was authenticated by human STR profiling and confirmed to be mycoplasma free. HepG2 cells were cultured in DMEM (Nanjing KeyGen Biotech Co., Ltd.) containing 10% fetal bovine serum (FBS; Biological Industries) and 1% penicillin-streptomycin (Nanjing KeyGen Biotech Co., Ltd.), and incubated in a humidified 5% CO2 incubator at 37°C.

The lentiviral vectors, GV513-ADAMTS16 (Ubi-MCS-CBh-gcGFP-IRES-puromycin) and GV358-BMP2 (Ubi-MCS-3FLAG-SV40-EGFP-IRES-puromycin), and the corresponding control lentiviruses containing the empty vector, CON335 and CON238, respectively, were purchased from Shanghai GeneChem Co., Ltd. Accession numbers for the two genes used in the present study are as follows: ADAMTS16, NM139056; BMP2, NM001200. For lentiviral infection, HepG2 cells were incubated with lentivirus at a MOI of 10 for 16 h, and the stable cell line was selected using 2 µg/ml puromycin for a week, followed by 2 µg/ml puromycin maintenance. The overexpression efficiency of ADAMTS16 and BMP2 were assessed through reverse transcription-quantitative PCR (RT-qPCR).

Transcriptome sequencing and analysis

In total, 40 HCC tissue samples were divided into TB-pos and TB-neg groups by two independent pathologists on the basis of hematoxylin-eosin (HE) staining for transcriptome sequencing, which was conducted by Berry Genomics Co., Ltd. Briefly, using the NEBNext® UltraTM RNA Library Prep Kit for Illumina® (E7770S, New England Biolabs, Inc.), total RNA (≥1 µg) from each sample was used for generating sequencing libraries in-line with the manufacturer's instructions. Oligo (dT) magnetic beads were used to enrich and purify poly A-containing mRNA, followed by random interruption of mRNA into short fragments, which served as templates for random-primed cDNA synthesis performed using reverse transcriptase. Subsequently, purified double-stranded cDNA was subjected to end-repair, A-tailing and adapter ligation. Moreover, AMPure XP beads (Beckman Coulter, Beverly, USA) were used to purify cDNA library fragments for selecting the 250-300 bp cDNA fragments. Lastly, PCR amplification was performed by ABI Q3TMReal-Time PCR System, followed by purification of PCR products using the AMPure XP beads for obtaining the cDNA library.

Following library construction, the Qubit 2.0 Fluorometer (Thermo Fisher Scientific, Inc.) was used to quantify the library before diluting to 1.5 ng/µl. Then, the Agilent 2100 Bioanalyzer was used to assess the library insert size. When the expected insert size was obtained, qPCR was conducted to precisely quantify library effective concentration (>2 nM) for ensuing library quality. After the library quality was confirmed, the Illumina platform was used for sequencing to generate 150 bp paired end reads.

The edgeR software package (version 3.28.1; http://bioconductor.org/packages/release/bioc/html/edgeR.html) (22) in the R/Bioconductor environment (Release 3.6.1) was used for differential expression analysis. To control the false discovery rate, the resulting P-values were adjusted according to the Benjamini and Hochberg's approach (23). Genes with log2 (Fold Change) >1 and Q<0.05 were assigned as differentially expressed. To annotate the function of these DEGs, Gene ontology (GO) enrichment analysis of DEG sets were implemented in topGO (version 2.38.1; http://www.bioconductor.org/packages/release/bioc/html/topGO.html) in the R/Bioconductor environment (Release 3.6.1). GO terms with adjusted P<0.05 were considered as significantly enriched by DEGs.

RT-qPCR

Total RNA from the HepG2 cells was extracted using TRIzol reagent (Ambion; Thermo Fisher Scientific, Inc.). Reverse transcription was conducted using the HiScript III RT SuperMix for qPCR (+gDNA wiper) (Vazyme Biotech Co., Ltd.) according to the manufacturer's protocol. The reverse transcription reactions were performed at 42°C for 2 min, followed by 37°C for 15 min and 85°C for 5 sec. qPCR was performed using ChamQ Universal SYBR qPCR Master Mix (Vazyme Biotech Co., Ltd.) in a CFX Connect Real-time System (Bio-Rad Laboratories Inc.). GAPDH was employed as the internal reference and the 2−∆∆Cq method was used for quantification (24). Primer sequences for GAPDH, ADAMTS16 and BMP2 were as follows: GAPDH, 5′-CTGACTTCAACAGCGACACC-3′ (forward) and 5′-TGCTGTAGCCAAATTCGTTGT-3′ (reverse); ADAMTS16, 5′-CCGGCCGGTACAAATTTTCG-3′ (forward) and 5′-AACAGCAGCTCCACAATCAGT-3′ (reverse); BMP2, 5′-AGAATGCAAGCAGGTGGGAA-3′ (forward) and 5′-TCTTGGTGCAAAGACCTGCT-3′ (reverse).

Inverse Matrigel invasion assays

The inverse Matrigel invasion assays were performed as reported previously (25). To evaluate the TB capacity of HepG2 cells, 8-µm pore sized Transwell chambers (Corning, Inc.) were used. Briefly, the Transwell chambers were hydrated using serum-free DMEM medium for 30 min. Then, the Matrigel (BD Biosciences) was added to each chamber and solidified in an incubator at 37°C for 1 h. Afterwards, the chambers were inverted and the cells (2×105/well) were seeded onto the upward facing underside of the chamber. Following cell attachment, serum-free DMEM medium was placed in the lower chamber and DMEM medium containing 20% FBS (serving as a chemoattractant) was added to the upper chamber. The cells were allowed to invade through the Matrigel for 3-5 days in a 37°C incubator. Finally, the 3D reconstruction of cell invasion was obtained through Zeiss 800 laser confocal microscopy. The ratio of the fluorescence values of GFP-positive budding cells/total fluorescence value of all GFP-positive cells × 100% was calculated using Image J software (version 1.8.0) to quantify the invaded cells (TB rate).

Spheroid-based sprouting assays

Spheroid-based sprouting assays were performed as previously described (26), with some optimization. HepG2 spheroids were obtained using the hanging drop method. HepG2 cells were suspended at a density of 1×104 cells/45 µl and then seeded on the lid of 48-well culture plates for 2 days in an incubator at 37°C. Then, all spheroids were harvested and embedded in Matrigel for 2 days in a 37°C incubator. Finally, images were captured using a light microscope (OPTIKA).

Immunohistochemistry (IHC)

In total, 308 formalin-fixed, paraffin-embedded tissue samples from patients with HCC were used to establish HCC tissue microarrays (TMAs) with a 2.0-mm diameter per core for IHC. In brief, TMAs were dewaxed, hydrated and antigen repaired by EDTA antigen repair buffer (PH 8.0), and the endogenous peroxidase activity was then blocked. TMAs were blocked in goat serum (OriGene Technologies, Inc.) at room temperature for 30 min to block non-specific staining. Primary antibodies were incubated with the TMAs overnight at 4°C, including ADAMTS16 (1:200; cat. no. DF9173; Affinity Biosciences) and BMP2 (1:200; cat. no. A0231; ABclonal Biotech Co., Ltd.). Subsequently, a horseradish peroxidase-labeled secondary antibody (KIT-5020; Maxim Co., Ltd., Fuzhou, China) was incubated with the TMAs for 30 min at room temperature. Finally, a chromogenic reaction was developed with DAB kit (DAB-0031 (20×); Fuzhou Maixin Biotech Co., Ltd.), followed by counterstaining with hematoxylin (G1120; Beijing Solarbio Science & Technology Co., Ltd.) for 10-30 sec at room temperature. All IHC staining analyses were assessed using a semi-quantitative scoring approach by two senior pathologists. IHC scores were calculated by multiplying staining intensity with the stained area, in which the staining intensity was scored as follows: 0 (no staining), 1 (light yellow staining), 2 (yellow-brown staining) and 3 (brown staining). The staining area was scored as follows: 1 (1–25%), 2 (26–50%), 3 (51–75%) and 4 (76–100%), according to the percentage of stained area in the field of vision. The median IHC score of ADAMTS16 or BMP2 was used as a cut-off to divide the samples into the low expression and high expression groups. ‘High’ was defined an IHC score higher than the cut-off value, and ‘low’ was defined as an IHC score lower than or equal the cut-off value.

Statistical analysis

SPSS 25.0 (IBM Corp.) was used for statistical analyses. Differences between two groups were analyzed using the Wilcoxon rank-sum test or unpaired student's t-test. The association of ADAMTS16 and BMP2 expressions with clinicopathological features was analyzed using the chi-square test or Fisher's exact test as appropriate. Kaplan-Meier (KM) curves were generated using the ‘survfit’ function in the survival package of R software (version 3.5.3), while significant differences in survival were compared using the log-rank test or Cramer-von Mises test as appropriate. Cramer-von Mises test was performed to generate the P-values when KM curves crossed over. P<0.05 was considered to indicate a statistically significant difference.

Results

Transcriptome sequencing analysis

A total of 40 surgical HCC specimens were divided into two groups: TB-pos group (n=21) and TB-neg group (n=19), followed by transcriptome sequencing analysis. As shown in the volcano plot in Fig. 1A, 245 DEGs including 95 upregulated DEGs and 150 downregulated DEGs were obtained for the TB-pos group compared with the TB-neg group. GO enrichment analysis was subsequently performed for the 95 upregulated DEGs. The top 20 upregulated biological processes (BPs) are shown in Fig. 1B. The predominant BPs of the upregulated DEGs are associated with embryonic kidney development.

Results of GO (BP) enrichment analysis for downregulated genes are shown in Fig. 1C. It was noted that downregulated genes were mainly involved in immune-related processes, such as immune response, innate immune response and response to type I interferon. Certain studies have illustrated that the downregulated immune-related genes are associated with tumor immune escape (2729).

The details of BP enrichment of upregulated genes in the GO analysis are provided in Table SI. In the present study, BPs of embryonic kidney development-related processes related to the upregulated genes of HCC with TB was made the focus as this is a novel area. Genes involved in the BPs associated with embryonic kidney development include ADAMTS16, BMP2, CALB1, FOXD1 and WT1. Furthermore, we found that there were 4 common upregulated genes in these embryonic kidney development-related processes, including ADAMTS16, BMP2, WT1 and FOXD1. ADAMTS16 and BMP2 were selected for further investigation. In addition, we noted that S100A9 which involved in neutrophil aggregation, was a poor prognosis-related gene. The survival analysis result of S100A9 based on TCGA data showed that the higher expression level of S100A9 was also linked with a worse prognosis of HCC patients (P=0.013) (Fig. S1). It was also reported in the literature that S100A9 expression could potentially serve as an independent prognostic marker for HCC (30).

High ADAMTS16 and BMP2 expression levels are associated with TB in HCC

To identify ADAMTS16 and BMP2 expression profiles within HCC tissues with different TB statuses, IHC staining was performed using TMAs of 308 paraffin-embedded HCC tissues. In addition, the DEGs of the budding cells and cancer center were compared. The statistical analysis results for the IHC staining scores of ADAMTS16 and BMP2 expression are summarized in Tables I and II. As shown in Table I, the staining scores of ADAMTS16 expression in the TB-pos HCC tissues were significantly higher than those in the TB-neg HCC tissues (P=0.005). However, the staining scores of ADAMTS16 expression demonstrated no significant difference between the tumor center and budding cells (P=0.174). As shown in Table II, the staining scores of BMP2 expression were significantly higher in TB-pos group compared with that in TB-neg group (P=0.015), and the significantly higher staining scores in budding cells than in tumor center (P=0.042) were observed. The representative results for IHC staining of ADAMTS16 and BMP2 expressions are illustrated in Fig. 2. These results demonstrated that upregulation of ADAMTS16 and BMP2 expression may be related to TB in HCC. Moreover, BMP2 expression was significantly increased in the budding cells compared with the tumor center.

Table I.

Association between a metalloproteinase domain with thrombospondin motifs 16 expression and TB in hepatocellular carcinoma tissues (n=308).

Table I.

Association between a metalloproteinase domain with thrombospondin motifs 16 expression and TB in hepatocellular carcinoma tissues (n=308).

Two-sample Wilcoxon rank-sum test

GroupMedian (P25, P75)Median difference (95% CI)Z valueP-value
TB −1.000 (−1.000-0.000)−2.8020.005a
  TB-neg2 (1, 4)
  TB-pos3 (1, 6)
Area 0.000 (−2.000-0.000)−1.3590.174
  Tumor center3 (2, 6)
  Budding cells4 (3, 8)

a P<0.05. neg, negative; pos, positive; TB, tumor budding; P25, lower quartile; P75, upper quartile.

Table II.

Association between bone morphogenetic protein 2 expression and TB in hepatocellular carcinoma tissues (n=308).

Table II.

Association between bone morphogenetic protein 2 expression and TB in hepatocellular carcinoma tissues (n=308).

Two-sample Wilcoxon rank-sum test

GroupMedian (P25, P75)Median difference (95% CI)Z valueP-value
TB 0.000 (0.000-0.000)−2.4340.015a
  TB-neg8 (8, 9)
  TB-pos8 (8, 12)
Area 0.000 (−2.000-0.000)−2.0380.042a
  Tumor center8 (8, 12)
  Budding cells12 (8, 12)

a P<0.05. neg, negative; pos, positive; TB, tumor budding, P25, lower quartile; P75, upper quartile.

ADAMTS16 and BMP2 promote the TB of liver cancer in vitro

To explore whether ADAMTS16 and BMP2 play a role in the TB of liver cancer, HepG2 cells with stable ADAMTS16-OE or BMP2-OE were constructed using lentiviral vectors. The overexpression efficiency was assessed through RT-qPCR assays (Fig. S2). Subsequently, the TB ability of HepG2 cells was evaluated using in vitro inverse Matrigel invasion and spheroid-based sprouting assays. As shown in Fig. 3A, the ratio of TB in ADAMTS16-OE or BMP2-OE HepG2 cells increased significantly relative to their respective controls. Similarly, the results of the spheroid-based sprouting assays demonstrated that ADAMTS16-OE or BMP2-OE in HepG2 cells resulted in more budding cells compared with the control cells (Fig. 3B). These results demonstrated that ADAMTS16 and BMP2 may be involved in the regulation of TB of liver cancer.

Association of ADAMTS16 and BMP2 expression levels with clinicopathological characteristics in HCC

To analyze the association of ADAMTS16 and BMP2 expression levels with HCC clinicopathological characteristics, 266 HCC cases with complete available clinicopathological information for analysis were enrolled. These 266 cases were classified into the low- or high-expression groups by considering the respective median IHC scores for ADAMTS16 and BMP2 expression as the cut-off. The relationship between the clinicopathological characteristics of patients and gene expression was assessed using the chi-square test and Fisher's exact test. Consequently, ADAMTS16 expression was found to be significantly associated with necrosis (P=0.023) and cholestasis (P=0.011) (Table III). As shown in Table IV, BMP2 expression level had a significant association with the Barcelona Clinic Liver Cancer (BCLC) stage (B-C vs. 0-A; P=0.003) and the vessels encapsulating tumor cluster (VETC; P=0.014), which is one of the vessel types in HCC.

Table III.

Association between ADAMTS16 expression and the clinicopathological characteristics in patients with hepatocellular carcinoma (n=266).

Table III.

Association between ADAMTS16 expression and the clinicopathological characteristics in patients with hepatocellular carcinoma (n=266).

ADAMTS16

Clinicopathological characteristicsLow, n (n=131)High, n (n=135)P-value
Age, years 0.848
  >604851
  ≤608384
Sex 0.359
  Male105114
  Female2621
HBV infection 0.193
  Positive116112
  Negative1523
HCV infection 0.122
  Positive04
  Negative131131
AFP serum level, ng/ml 0.407
  >4004338
  ≤4008897
Liver cirrhosis (Yes vs. No) 0.359
  Yes106103
  No2532
BCLC stage 0.325
  B-C2116
  0-A110119
Tumor size, cm 0.969
  >55557
  ≤57678
Tumor number 0.114
  Multiple1221
  Single119114
Intrahepatic metastasis 0.114
  Yes1221
  No119114
Collective invasion 0.693
  Yes911
  No122124
Ki-67, % 0.236
  >303124
  ≤30100111
Necrosis 0.023a
  Yes2815
  No103120
Vessel carcinoma embolus 0.285
  Yes1712
  No114123
Microtrabecular pattern 0.735
  Yes106107
  No2528
Macrotrabecular pattern 0.780
  Yes7575
  No5660
Pseudoglandular pattern 0.130
  Yes2334
  No108101
Compact pattern 0.865
  Yes5356
  No7879
Cholestasis 0.011a
  Yes1734
  No114101
Hyaline bodies 0.273
  Yes2331
  No108104
Steatosis 0.908
  Yes2424
  No107111
Edmondson grade 0.136
  III–IV4636
  I–II8599
VETC 0.497
  Yes3339
  No9896

a P<0.05. ADAMTS16, a metalloproteinase domain with thrombospondin motifs 16; HBV, hepatitis B virus; HCV, hepatitis C virus; AFP, α-fetoprotein; BCLC, Barcelona Clinic Liver Cancer; VETC, vessels-encapsulate tumor cluster.

Table IV.

Association between BMP2 expression and the clinicopathological characteristics in patients with hepatocellular carcinoma (n=266).

Table IV.

Association between BMP2 expression and the clinicopathological characteristics in patients with hepatocellular carcinoma (n=266).

BMP2

Clinicopathological characteristicsLow, n (n=172)High, n (n=94)P-value
Age, years 0.290
  >606831
  ≤6010463
Sex 0.588
  Male14079
  Female3215
HBV infection 0.105
  Positive14385
  Negative299
HCV infection 0.301
  Positive40
  Negative16894
AFP serum level, ng/ml 0.508
  >4005031
  ≤40012263
Liver cirrhosis (Yes vs. No) 0.789
  Yes13673
  No3621
BCLC stage 0.003a
  B-C1621
  0-A15673
Tumor size, cm 0.054
  >56547
  ≤510747
Tumor number 0.091
  Multiple1716
  Single15578
Intrahepatic metastasis 0.091
  Yes1716
  No15578
Collective invasion 0.604
  Yes146
  No15888
Ki-67, % 0.417
  >303322
  ≤3013972
Necrosis 0.530
  Yes2617
  No14677
Vessel carcinoma embolus 0.123
  Yes1514
  No15780
Microtrabecular pattern 0.683
  Yes13974
  No3320
Macrotrabecular pattern 0.302
  Yes9357
  No7937
Pseudoglandular pattern 0.964
  Yes3720
  No13574
Compact pattern 0.511
  Yes7336
  No9958
Cholestasis 0.750
  Yes3219
  No14075
Hyaline bodies 0.352
  Yes3222
  No14072
Steatosis 0.186
  Yes3513
  No13781
Edmondson grade 0.264
  III–IV4933
  I–II12361
VETC 0.014a
  Yes3834
  No13460

a P<0.05. BMP2, bone morphogenetic protein 2; HBV, hepatitis B virus; HCV, hepatitis C virus; AFP, α-fetoprotein; BCLC, Barcelona Clinic Liver Cancer; VETC, vessels-encapsulate tumor cluster.

Relationship between ADAMTS16 and BMP2 levels and the prognosis of patients with HCC

A total of 242 HCC cases with available follow-up data were enrolled for survival analysis. These 242 HCC cases were divided into two groups (low or high expression) based on the median IHC scores for ADAMTS16 or BMP2. As shown in Fig. 4A and B, the KM survival curves demonstrated that ADAMTS16 expression was statistically significantly associated with overall survival (OS; ADAMTS16, P=0.046) in patients with HCC. In addition, BMP2 expression was not associated with the OS of patients with HCC (BMP2, P=0.59). Subsequently, the survival curves of these two genes were downloaded from the GEPIA database (http://gepia.cancer-pku.cn/detail.php). As shown in Fig. 4C and D, the survival time of patients with HCC with high BMP2 expression significantly decreased compared with that of patients with low BMP2 expression (P=0.0081). ADAMTS16 expression did not differ significantly between the two groups (P=0.24). The GEO database (accession no. GSE76427; http://www.ncbi.nlm.nih.gov/geo/) was also utilized to perform survival analyses comparing expression levels of ADAMTS16 and BMP2 in HCC. The results demonstrated that neither ADAMTS16 (P=0.28) nor BMP2 (P=0.61) expression were associated with the OS time of patients with HCC (Fig. 4E and F). The potential reasons for this discrepancy were elucidated in the discussion.

Discussion

TB, a histological phenomenon observed in the tumor invasive edge, has been recognized as the initial stage of cancer invasion and metastasis (31,32). TB has been reported to portend lymphatic/vascular invasion, lymph node metastasis, distant metastasis, failure to respond to neoadjuvant chemoradiotherapy, locoregional relapse and poor survival (6,7,3337). Certain studies have demonstrated an association between TB and low survival rates in numerous types of solid cancers, including nasopharyngeal, lung, pancreatic and esophageal cancer (69). However, little is known about the effect of TB on HCC. Moreover, TB can only be definitively determined following surgery and based on detailed pathological and immunohistochemical examinations, thus limiting its role in selecting the therapeutic options for HCC. Therefore, exploring the role of TB in HCC and its molecular mechanisms is crucial for identifying new therapeutic targets for combating HCC.

In the present study, the expression profiles of 40 HCC tissues were analyzed using transcriptome sequencing technology. Functional annotation indicated that upregulated DEGs, obtained by comparing the gene expression of TB-pos and TB-neg HCC tissues, were mainly involved in embryonic kidney development, including nephron tubule development, kidney morphogenesis, renal tubule development and ureteric bud development. With advancements in developmental and cancer biology, numerous studies have suggested that cancer invasion and metastasis are similar to normal embryonic development (3840). Hence, the TB process may be considered to simulate embryonic kidney development. Furthermore, ADAMTS16 and BMP2 were selected from the enriched genes associated with embryonic kidney development for follow-up studies. ADAMTS16 belongs to the ADAMTS family and it has been demonstrated to promote the proliferation and invasion of gastric cancer (41) and esophageal cancer (15) cells. However, to the best of our knowledge, the function of ADAMTS16 has not yet been studied in HCC. BMP2, which belongs to the transforming growth factor-β superfamily, participates in different cancer occurrence and development processes (1719). Certain studies have indicated that BMP2 may enhance HCC cell growth, invasion and migration (20,21). However, to the best of our knowledge, no study has discussed the relationship between BMP2 and TB in HCC.

The IHC results in the present study indicated an upregulation of ADAMTS16 and BMP2 expression in TB-pos HCC tissues compared with TB-neg HCC tissues. BMP2 expression in budding cells was also increased compared with that in the tumor center. Moreover, through the inverse Matrigel invasion and spheroid-based sprouting assays, the present study revealed that ADAMTS16 and BMP2 may regulate the TB process of liver cancer. Therefore, we hypothesize that these two genes may be associated with the malignant progression of liver cancer. Furthermore, the association of ADAMTS16 and BMP2 expression with clinicopathological characteristics of patients with HCC was analyzed, and an association between ADAMTS16 expression and necrosis and cholestasis was confirmed in. In addition, BMP2 expression was significantly associated with the BCLC stage and VETC.

It is well known that rapidly growing tumor cells, which represent an important malignant behavior of tumors, require an adequate supply of oxygen and nutrients, and the blood supply cannot meet this need of rapid tumor growth, eventually resulting in tumor necrosis (42). Therefore, the significant association between ADAMTS16 and necrosis in HCC observed in the present study may be caused by the ability of ADAMTS16 to promote tumor malignant progression of HCC. To the best of our knowledge, no study has reported the association of ADAMTS family members with cholestasis. However, a close relative of the ADAMTS family, ADAM17, has been reported to be related to cholestasis (43). Increased ADAM17 expression was found in patients with two important cholestatic liver diseases, including primary biliary cholangitis and primary sclerosing cholangitis (43). Given the functional similarity of these two genes, the result regarding the association of ADAMTS16 with cholestasis in HCC is reasonable and reliable.

As for BMP2, in the present study, patients with BCLC stage B-C had significantly higher BMP2 expression levels than patients with BCLC stage 0-A, suggesting the involvement of BMP2 in the progression and metastasis of HCC. Previous studies have demonstrated that BMP2 may promote HCC cell proliferation and invasion, thereby promoting malignant progression of HCC (20,21,44). As such, the results of the present study are in accordance with previously reported results. Certain studies have also demonstrated that BMP2 promotes angiogenesis of solid tumors, including HCC (45,46). In addition, VETC, a novel vascular pattern distinct from microvascular invasion, has become a powerful predictor of aggressive HCC (47). To the best of our knowledge, the present study is the first to report that BMP2 is significantly associated with VETC in HCC, which may explain the mechanism of this specific angiogenesis pattern in HCC. Tumor malignancy is closely associated with the invasiveness and metastasis of tumor which depends upon EMT (48,49). And TB is considered to be an EMT-like process (10). It has been documented that ADAMTS16 may promote cell migration and invasion through the NF-κB pathway (41). In addition, BMP2 also enhances the migration, invasion and EMT of tumor cells through the m-TOR signaling pathway (45,50). Hence, it is speculated that a contributing mechanism to TB in HCC may involve the m-TOR and/or NF-κB pathways.

The overall survival analysis of 242 patients with HCC indicated that ADAMTS16 expression was associated with HCC prognosis whereas BMP2 expression was not associated with HCC prognosis. In addition, the survival data obtained in the present study is inconsistent with the results predicted using the GSE76427 dataset and the results predicted using GEPIA, which is a The Cancer Genome Atlas (TCGA)-based online tool. The results of the GEPIA analysis demonstrated that BMP2 upregulation was significantly associated with poor OS. The results of the GSE76427 dataset showed that neither ADAMTS16 expression nor BMP2 expression was associated with HCC prognosis. The possible reasons for this discrepancy are as follows: Firstly, sample size varied widely across these three cohorts. which may cause the inconsistent results. And compared with the large sample size within the TCGA cohort, the retrospective cohort of the present study and the GSE76427 cohort have relatively small sample sizes. Secondly, the sources of the tumor samples in the three cohorts were different. The majority of patients from TCGA database are white. In GSE76427 cohort, all of the patients were derived from Singapore. And our study is based on data collected from a single center (Affiliated Hospital of Jining Medical University, China). Thirdly, as patient characteristics, surgical skills and treatment regimens are different among countries, the final outcome of patients with HCC could be affected. The absence of animal models of HCC is also a limitation of this study, animal models of HCC will be constructed for further study validation in the future.

Regardless of these limitations, to the best of our knowledge, the present study was the first to investigate the TB-related molecular mechanism in HCC. The findings of the present study provide evidence for mechanism studies of TB. Moreover, the present study provides a basis for the potential application of ADAMTS16 and BMP2 as predictive diagnosis markers and treatment targets for HCC.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was funded by the National Natural Science Foundation of China (grant no. 81972629), the Taishan Scholars Program of Shandong Province (grant no. tsqn201909193), the Shandong Youth Innovation and Technology program (grant no. 2020KJL003), the Jining Research and Development Program (grant nos. 2021YXNS065 and 2021YXNS075), and the Research Fund for Lin He Academician New Medicine (grant no. JYHL2021FMS12).

Availability of data and materials

The RNA-Seq datasets generated and/or analyzed during the current study are available in the GEO repository, the accession number is GSE227335. All other data generated or analyzed during this study are included in this published article.

Authors' contributions

DJ performed the experiments, data analysis and collection of clinical specimens, and drafted the manuscript. SX assisted with the data analysis and reverse transcription-quantitative PCR. CZ performed the bioinformatics analysis of the GEO database. CZ, CH and LL contributed to the analysis and interpretation of the data. MZ and HW contributed to the acquisition and interpretation of the data. DY and YL conceived and designed the study. YL revised the manuscript. DJ and SX confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Written informed consent was obtained from all patients. The study was approved by The Ethics Committee of the Affiliated Hospital of Jining Medical University (ethical approval no. 2021C145; Jining, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Yang JD, Hainaut P, Gores GJ, Amadou A, Plymoth A and Roberts LR: A global view of hepatocellular carcinoma: Trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol. 16:589–604. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Turajlic S and Swanton C: Metastasis as an evolutionary process. Science. 352:169–175. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Hase K, Shatney C, Johnson D, Trollope M and Vierra M: Prognostic value of tumor ‘budding’ in patients with colorectal cancer. Dis Colon Rectum. 36:627–635. 1993. View Article : Google Scholar : PubMed/NCBI

6 

Koike M, Kodera Y, Itoh Y, Nakayama G, Fujiwara M, Hamajima N and Nakao A: Multivariate analysis of the pathologic features of esophageal squamous cell cancer: Tumor budding is a significant independent prognostic factor. Ann Surg Oncol. 15:1977–1982. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Luo WR, Gao F, Li SY and Yao KT: Tumour budding and the expression of cancer stem cell marker aldehyde dehydrogenase 1 in nasopharyngeal carcinoma. Histopathology. 61:1072–1081. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Masuda R, Kijima H, Imamura N, Aruga N, Nakamura Y, Masuda D, Takeichi H, Kato N, Nakagawa T, Tanaka M, et al: Tumor budding is a significant indicator of a poor prognosis in lung squamous cell carcinoma patients. Mol Med Rep. 6:937–943. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Karamitopoulou E, Zlobec I, Born D, Kondi-Pafiti A, Lykoudis P, Mellou A, Gennatas K, Gloor B and Lugli A: Tumour budding is a strong and independent prognostic factor in pancreatic cancer. Eur J Cancer. 49:1032–1039. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Grigore AD, Jolly MK, Jia D, Farach-Carson MC and Levine H: Tumor budding: The name is EMT. Partial EMT. J Clin Med. 5:512016. View Article : Google Scholar : PubMed/NCBI

11 

De Smedt L, Palmans S, Andel D, Govaere O, Boeckx B, Smeets D, Galle E, Wouters J, Barras D, Suffiotti M, et al: Expression profiling of budding cells in colorectal cancer reveals an EMT-like phenotype and molecular subtype switching. Br J Cancer. 116:58–65. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Kairaluoma V, Kemi N, Pohjanen VM, Saarnio J and Helminen O: Tumour budding and tumour-stroma ratio in hepatocellular carcinoma. Br J Cancer. 123:38–45. 2020. View Article : Google Scholar : PubMed/NCBI

13 

Wei L, Delin Z, Kefei Y, Hong W, Jiwei H and Yange Z: A classification based on tumor budding and immune score for patients with hepatocellular carcinoma. Oncoimmunology. 9:16724952019. View Article : Google Scholar : PubMed/NCBI

14 

Zhang D, Qian C, Wei H and Qian X: Identification of the prognostic value of tumor microenvironment-related genes in esophageal squamous cell carcinoma. Front Mol Biosci. 7:5994752020. View Article : Google Scholar : PubMed/NCBI

15 

Sakamoto N, Oue N, Noguchi T, Sentani K, Anami K, Sanada Y, Yoshida K and Yasui W: Serial analysis of gene expression of esophageal squamous cell carcinoma: ADAMTS16 is upregulated in esophageal squamous cell carcinoma. Cancer Sci. 101:1038–1044. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Mersakova S, Janikova K, Kalman M, Marcinek J, Grendar M, Vojtko M, Kycina R, Pindura M, Janik J, Mikolajcik P, et al: Cancer stem cell marker expression and methylation status in patients with colorectal cancer. Oncol Lett. 24:2312022. View Article : Google Scholar : PubMed/NCBI

17 

Raida M, Clement JH, Ameri K, Han C, Leek RD and Harris AL: Expression of bone morphogenetic protein 2 in breast cancer cells inhibits hypoxic cell death. Int J Oncol. 26:1465–1470. 2005.PubMed/NCBI

18 

Yang S, Pham LK, Liao CP, Frenkel B, Reddi AH and Roy-Burman P: A novel bone morphogenetic protein signaling in heterotypic cell interactions in prostate cancer. Cancer Res. 68:198–205. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Hsu YL, Huang MS, Yang CJ, Hung JY, Wu LY and Kuo PL: Lung tumor-associated osteoblast-derived bone morphogenetic protein-2 increased epithelial-to-mesenchymal transition of cancer by Runx2/Snail signaling pathway. J Biol Chem. 286:37335–37346. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Wu G, Huang F, Chen Y, Zhuang Y, Huang Y and Xie Y: High levels of BMP2 promote liver cancer growth via the activation of myeloid-derived suppressor cells. Front Oncol. 10:1942020. View Article : Google Scholar : PubMed/NCBI

21 

Liu Z, Sun J, Wang X and Cao Z: MicroRNA-129-5p promotes proliferation and metastasis of hepatocellular carcinoma by regulating the BMP2 gene. Exp Ther Med. 21:2572021. View Article : Google Scholar : PubMed/NCBI

22 

Robinson MD, McCarthy DJ and Smyth GK: edgeR: A bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 26:139–140. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Benjamini Y and Hochberg Y: Controlling the false discovery rate-a practical and powerful approach to multiple testing. J Roy Statist Soc B. 57:289–300. 1995.

24 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Scott RW, Crighton D and Olson MF: Modeling and imaging 3-dimensional collective cell invasion. J Vis Exp. 35252011.PubMed/NCBI

26 

Pfisterer L and Korff T: Spheroid-based in vitro angiogenesis model. Methods Mol Biol. 1430:167–177. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Muenst S, Läubli H, Soysal SD, Zippelius A, Tzankov A and Hoeller S: The immune system and cancer evasion strategies: Therapeutic concepts. J Intern Med. 279:541–562. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Aust S, Felix S, Auer K, Bachmayr-Heyda A, Kenner L, Dekan S, Meier SM, Gerner C, Grimm C and Pils D: Absence of PD-L1 on tumor cells is associated with reduced MHC I expression and PD-L1 expression increases in recurrent serous ovarian cancer. Sci Rep. 7:429292017. View Article : Google Scholar : PubMed/NCBI

29 

Baloche V, Rivière J, Tran TBT, Gelin A, Bawa O, Signolle N, Diop MBK, Dessen P, Beq S, David M and Busson P: Serial transplantation unmasks galectin-9 contribution to tumor immune escape in the MB49 murine model. Sci Rep. 11:52272021. View Article : Google Scholar : PubMed/NCBI

30 

Liao J, Li JZ, Xu J, Xu Y, Wen WP, Zheng L and Li L: High S100A9+ cell density predicts a poor prognosis in hepatocellular carcinoma patients after curative resection. Aging (Albany NY). 13:16367–16380. 2021. View Article : Google Scholar : PubMed/NCBI

31 

Liang F, Cao W, Wang Y, Li L, Zhang G and Wang Z: The prognostic value of tumor budding in invasive breast cancer. Pathol Res Pract. 209:269–275. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Prall F: Tumour budding in colorectal carcinoma. Histopathology. 50:151–162. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Morodomi T, Isomoto H, Shirouzu K, Kakegawa K, Irie K and Morimatsu M: An index for estimating the probability of lymph node metastasis in rectal cancers. Lymph node metastasis and the histopathology of actively invasive regions of cancer. Cancer. 63:539–543. 1989. View Article : Google Scholar : PubMed/NCBI

34 

Ueno H, Murphy J, Jass JR, Mochizuki H and Talbot IC: Tumour ‘budding’ as an index to estimate the potential of aggressiveness in rectal cancer. Histopathology. 40:127–132. 2002. View Article : Google Scholar : PubMed/NCBI

35 

Mitrovic B, Schaeffer DF, Riddell RH and Kirsch R: Tumor budding in colorectal carcinoma: Time to take notice. Mod Pathol. 25:1315–1325. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Satoh K, Nimura S, Aoki M, Hamasaki M, Koga K, Iwasaki H, Yamashita Y, Kataoka H and Nabeshima K: Tumor budding in colorectal carcinoma assessed by cytokeratin immunostaining and budding areas: Possible involvement of c-Met. Cancer Sci. 105:1487–1495. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Rogers AC, Gibbons D, Hanly AM, Hyland JM, O'Connell PR, Winter DC and Sheahan K: Prognostic significance of tumor budding in rectal cancer biopsies before neoadjuvant therapy. Mod Pathol. 27:156–162. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Li A and Machesky LM: Melanoblasts on the move: Rac1 sets the pace. Small GTPases. 3:115–119. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Gupta S and Maitra A: EMT: Matter of life or death? Cell. 164:840–842. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Yu M, Bardia A, Wittner BS, Stott SL, Smas ME, Ting DT, Isakoff SJ, Ciciliano JC, Wells MN, Shah AM, et al: Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science. 339:580–584. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Li T, Zhou J, Jiang Y, Zhao Y, Huang J, Li W, Huang Z, Chen Z, Tang X, Chen H and Yang Z: The novel protein ADAMTS16 promotes gastric carcinogenesis by targeting IFI27 through the NF-κb signaling pathway. Int J Mol Sci. 23:110222022. View Article : Google Scholar : PubMed/NCBI

42 

Wu CX, Lin GS, Lin ZX, Zhang JD, Chen L, Liu SY, Tang WL, Qiu XX and Zhou CF: Peritumoral edema on magnetic resonance imaging predicts a poor clinical outcome in malignant glioma. Oncol Lett. 10:2769–2776. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Almishri W, Swain LA, D'Mello C, Le TS, Urbanski SJ and Nguyen HH: ADAM metalloproteinase domain 17 regulates cholestasis-associated liver injury and sickness behavior development in mice. Front Immunol. 12:7791192022. View Article : Google Scholar : PubMed/NCBI

44 

Guo J, Guo M and Zheng J: Inhibition of BONE MORPHOGENETIC PROtein 2 suppresses the stemness maintenance of cancer stem cells in hepatocellular carcinoma via the MAPK/ERK pathway. Cancer Manag Res. 13:773–785. 2021. View Article : Google Scholar : PubMed/NCBI

45 

Zuo WH, Zeng P, Chen X, Lu YJ, Li A and Wu JB: Promotive effects of bone morphogenetic protein 2 on angiogenesis in hepatocarcinoma via multiple signal pathways. Sci Rep. 6:374992016. View Article : Google Scholar : PubMed/NCBI

46 

Raida M, Clement JH, Leek RD, Ameri K, Bicknell R, Niederwieser D and Harris AL: Bone morphogenetic protein 2 (BMP-2) and induction of tumor angiogenesis. J Cancer Res Clin Oncol. 131:741–750. 2005. View Article : Google Scholar : PubMed/NCBI

47 

Renne SL, Woo HY, Allegra S, Rudini N, Yano H, Donadon M, Viganò L, Akiba J, Lee HS, Rhee H, et al: Vessels encapsulating tumor clusters (VETC) is a powerful predictor of aggressive hepatocellular carcinoma. Hepatology. 71:183–195. 2020. View Article : Google Scholar : PubMed/NCBI

48 

Rodriguez MI, Peralta-Leal A, O'Valle F, Rodriguez-Vargas JM, Gonzalez-Flores A, Majuelos-Melguizo J, López L, Serrano S, de Herreros AG, Rodríguez-Manzaneque JC, et al: PARP-1 regulates metastatic melanoma through modulation of vimentin-induced malignant transformation. PLoS Genet. 9:e10035312013. View Article : Google Scholar : PubMed/NCBI

49 

Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, et al: The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 133:704–715. 2008. View Article : Google Scholar : PubMed/NCBI

50 

Wang MH, Zhou XM, Zhang MY, Shi L, Xiao RW, Zeng LS, Yang XZ, Zheng XFS, Wang HY and Mai SJ: BMP2 promotes proliferation and invasion of nasopharyngeal carcinoma cells via mTORC1 pathway. Aging (Albany NY). 9:1326–1340. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2023
Volume 25 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jiang D, Xu S, Zhang C, Hu C, Li L, Zhang M, Wu H, Yang D and Liu Y: Association between the expression levels of ADAMTS16 and BMP2 and tumor budding in hepatocellular carcinoma. Oncol Lett 25: 256, 2023
APA
Jiang, D., Xu, S., Zhang, C., Hu, C., Li, L., Zhang, M. ... Liu, Y. (2023). Association between the expression levels of ADAMTS16 and BMP2 and tumor budding in hepatocellular carcinoma. Oncology Letters, 25, 256. https://doi.org/10.3892/ol.2023.13842
MLA
Jiang, D., Xu, S., Zhang, C., Hu, C., Li, L., Zhang, M., Wu, H., Yang, D., Liu, Y."Association between the expression levels of ADAMTS16 and BMP2 and tumor budding in hepatocellular carcinoma". Oncology Letters 25.6 (2023): 256.
Chicago
Jiang, D., Xu, S., Zhang, C., Hu, C., Li, L., Zhang, M., Wu, H., Yang, D., Liu, Y."Association between the expression levels of ADAMTS16 and BMP2 and tumor budding in hepatocellular carcinoma". Oncology Letters 25, no. 6 (2023): 256. https://doi.org/10.3892/ol.2023.13842