Open Access

Impact of JMJD6 on intrahepatic cholangiocarcinoma

  • Authors:
    • Yukiko Kosai-Fujimoto
    • Shinji Itoh
    • Kyohei Yugawa
    • Takasuke Fukuhara
    • Daisuke Okuzaki
    • Takeo Toshima
    • Noboru Harada
    • Yoshinao Oda
    • Tomoharu Yoshizumi
    • Masaki Mori
  • View Affiliations

  • Published online on: June 23, 2022     https://doi.org/10.3892/mco.2022.2564
  • Article Number: 131
  • Copyright: © Kosai-Fujimoto 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

The association of Jumonji domain‑containing 6 (JMJD6) with the prognosis of various types of cancer has been demonstrated, except in intrahepatic cholangiocarcinoma (ICC). The present study aimed to clarify the impact of JMJD6 on ICC. The liver specimens of 51 patients who underwent surgery for ICC were analyzed for JMJD6 expression using immunohistochemistry staining. The relationship between clinicopathological factors and JMJD6 expression was investigated. The cellular activity was also evaluated in JMJD6 knocked down cells with Transwell migration assay and viability assay. In the immunohistochemistry staining of clinical samples, high expression of JMJD6 was seen in 32 of 51 samples. High expression was also associated with improved overall survival (OS) and recurrence‑free survival (RFS) (P=0.0033 and 0.048, respectively). Further analyses revealed that higher JMJD6 expression was one of the improved independent prognostic factors of OS and RFS. Expression of JMJD6 was knocked down in commercial culture cell lines of ICC, and RNA and protein were extracted to analyze the downstream gene expression using RNA‑sequencing and western blotting. JMJD6 knockdown was associated with higher programmed death‑ligand 1 (PD‑L1) expression in RNA‑sequencing and western blotting. In addition, PD‑L1 expression was higher in JMJD6 low expression clinical samples when measured using immunohistochemistry staining. In conclusion, high expression of JMJD6 was an independent favorable prognostic factor of ICC. JMJD6 may influence the prognosis of ICC through the regulation of PD‑L1 expression.

Introduction

Intrahepatic cholangiocarcinoma (ICC) is the second most common primary hepatic malignancy, accounting for >5% of primary liver cancers, and the number of cases is increasing worldwide (1). The only potentially curative treatment of ICC is surgical resection, although some patients subsequently develop recurrence (2). Several combinations of systemic chemotherapy have shown to improve patients' survival; however, the etiology and pathogenesis of ICC remain poorly understood (3-5).

Jumonji domain-containing 6 (JMJD6), a member of the Jumonji C domain-containing family of proteins, was originally identified as a phosphatidylserine receptor (PSR) on cell surface (6). Subsequent studies have demonstrated that JMJD6 is located in the nucleus and has demethylase and hydroxylase activities toward histone and non-histone proteins (7,8). There is growing evidence to indicate that JMJD6 overexpression is associated with advanced clinicopathological stage, increased aggressiveness, and poor survival in various types of cancer; however, the impact of JMJD6 on ICC has not been reported yet (9).

Tumor immunology is a hot topic describing the interaction between the immune system and tumor cells. Understanding these interactions is important for the development of novel therapies for cancer. Immune checkpoint inhibitors function by reducing the suppression of T cells, especially CD8+ T cells, to improve tumor-specific responses (10,11). An increasing number of reports describe the relationship between CD8+ T cells and ICC prognosis. Our institution has shown that decreased microvessel density is related to worse prognosis, and Asahi et al also reported that high CD8 count could be an improved prognostic factor of ICC (12,13).

In this study, we assessed the clinical relevance and prognostic significance of JMJD6 expression in ICC. We also aimed to reveal the possible mechanism and the relationship between JMJD6 and the tumor immunological environment via in vitro JMJD6 knockdown studies.

Materials and methods

Patients and tumor samples

We retrospectively examined patients with primary ICC who underwent surgical resection at the Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University. Fifty-three patients with ICC who were diagnosed between May 1998 and August 2017 were eligible for inclusion in the study. The patients provided the written consent for the use of their tissues for future scientific research at the time of collection. Paraffin-embedded specimens were retrieved from the Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University. The analyzed clinicopathological features included the age at surgery, sex, pathological stage (8th edition AJCC/UICC staging manual), microvascular invasion, laboratory data, and the clinical course of each patient (14). This study was approved by the Clinical Research Ethics Committee of Kyushu University Hospital according to the Ethical Guidelines of the Japanese Government (approval no. 30-578).

Immunohistochemistry (IHC)

Formalin-fixed, paraffin-embedded tissue sections from patients with ICC were immunostained for JMJD6 (PSR H-7, sc-28348, Santa Cruz Biotechnology, Inc.; 1:100) and PD-L1 (clone SP142, Spring Bioscience; 1:100). Positive JMJD6 nuclear expression was defined as nuclear staining of ≥40% (Fig. 1). The programmed death-ligand 1 (PD-L1) expression on the membrane and cytoplasm was defined as positive when the percentage of positive cells was ≥5% of ICC cells (15). Immunohistochemical evaluations were independently performed by two observers (Y.K. and K.Y.) who were blinded to the clinical backgrounds of the patients. If the difference between evaluations was >10%, the evaluations were repeated. The findings of the two observers were averaged and considered final. The capture of microscopic images and quantitative analyses were undertaken on the NanoZoomer platform (Hamamatsu Photonics), and we ensured that the results matched with the observers' results.

Cell lines

The cholangiocarcinoma cell lines, SSP-25 and HuH-28, were obtained from Riken Bioresource Center, Tsukuba, Japan. The cell lines were originally isolated from intrahepatic cholangiocarcinoma specimens obtained from surgical resection of Japanese adult patients. The SSP-25 cell line was authenticated by STR profiling (supplemental document). The cells were incubated at 37˚C and 5% CO2 in RPMI media (Thermo Fisher Scientific, Inc.) supplemented with heat-inactivated 10% fetal bovine serum and penicillin-streptomycin.

JMJD6 siRNA transfection

The siRNA transfection was performed as previously described (16). Lipofectamine RNAiMAX (Invitrogen; Thermo Fisher Scientific, Inc.) was used to transfect the cells with siRNA (17). The siRNAs were obtained from Dharmacon, Inc. (siJMJD6-1 CAT# J-010363-10-0002, sequence=GGAGAGCACUCGAGAUGAU, siJMJD6-2 CAT# J-010363-12-0002, sequence=GGUAUAGGAUUUUGAAGCA, Control (non-targeting pool) CAT# D-001810-10-05). To facilitate transfection, the cells per well were incubated in 10% FBS containing RPMI; subsequently, they were plated to 40% confluence on a 6-well plate during transfection. We mixed 150 µl of Opti-MEM and 9 µl of RNAiMAX and subjected the mixture to incubation for 5 min at room temperature. In another tube, 10 pM siRNA in 3 ml of Opti-MEM and 150 µl of Opti-MEM were combined. Subsequently, we added siRNA solution to the diluted RNAiMAX reagent, and 250 µl of the prepared siRNA/RNAiMAX mixtures per well was used for incubation at room temperature for 25 min. Afterward, the 2.5x105 cells per well and the solution were combined. They were incubated for 6 h at 37˚C, and the mixture was replaced with RPMI with 10% FBS. The cells were incubated for 72 h in maximum, and the transfection efficiency was monitored every 24 h using western blotting and real-time polymerase chain reaction (PCR) in order to optimize the appropriate incubation time, and was decided to be 48 h.

Transwell migration assay and viability assay

Transwell migration assay: A total of 4x104 SSP-25 cells resuspended in 250 µl RPMI were placed on an 8.0-µm Transparent PET Membrane (Corning Inc.). The chamber was placed in a 24-well plate containing 750 µl RPMI and 10% FBS. After incubation at 37˚C overnight, migrating cells were stained with Diff-Quik (KACLaS) and were counted manually in five random microscopic fields at x200 magnification and quantified using ImageJ software (https://imagej.net/). The viability of the cells was examined by the CellTiter-Glo (CTG) assay (Promega) according to the manufacturer's instructions. Briefly, the cells were plated in 96-well plates with enough number of cells to 100% confluent in 24 h. The luminescence was read and quantified in 24 h with Multiskan GO spectrophotometric microplate reader (Thermo Fisher Scientific, Inc.).

RNA extraction and sequencing

RNA was extracted using the Maxwell(R) RSC simplyRNA tissue kit (Promega). Whole transcriptome sequencing was applied to RNA samples using the Illumina HiSeq 3000 platform in a 100-bp single-end mode. Sequenced reads were mapped to the human reference genome sequence (hg19) using TopHat version 2.0.13 in combination with Bowtie 2 version 2.2.3 and SAMTools version 1.0. The number of fragments per kilobase of exon per million mapped fragments was calculated using Cuffnorm version 2.2.1. RNA-Seq data were calculated as the fold change between samples with two-tailed Student's t-test (P<0.1) using the Subio Platform and Subio Basic Plug-in (v1.20; Subio, Inc.). Thresholds were set at a fold change of 2.0 and P-values of <0.05. Raw data of this study were submitted to Gene Expression Omnibus (accession no. GSE171974).

Reverse transcription-quantitative PCR (RT-qPCR)

One microgram of total RNA was converted to cDNA using the SuperScript III First-Strand Synthesis Supermix (Thermo Fisher Scientific, Inc.) with oligo-dT primers as per manufacturer's instructions. Quantification was determined using the ΔΔCt method relative to a β-actin control. The qRT-PCR primers used were as follows: JMJD6 Hs00397095_m1 and β-actin Hs01060665_g1. Assays were performed using the TaqMan Fast Advanced Master Mix (Thermo Fisher Scientific, Inc.) (18,19).

Western blot assay

Western blotting was performed as previously described (19). Briefly, whole-cell lysis was performed in RIPA buffer containing protease inhibitors (Nacalai Tesque). Proteins were separated by polyacrylamide gel electrophoresis and transferred to polyvinylidene difluoride membranes. After blocking with blocking buffer supplied in the iBind Western System (Thermo Fisher Scientific, Inc.), membranes were incubated with the primary antibody. Monoclonal antibodies for PD-L1 (1:1,000, E1L3N; Cell Signaling Technology), JMJD6 (1:200; Santa Cruz Biotechnology, Inc.), and β-actin (1:1,000; Cell Signaling Technology) were used. The JMJD6 antibody was derived from mouse, and the other antibodies were derived from rabbit. Horseradish peroxidase-conjugated secondary antibodies for mouse (1:1,000; Abcam) and rabbit (1:2,000; Abcam) were used. Primary and secondary antibodies were simultaneously applied on the rockers of the iBind Western System after setting membranes on a paper filter. Antibody binding was detected by enhanced chemiluminescence assays, and each band was detected using an Amersham Imager 600 (GE Healthcare Life Sciences).

Statistical analysis

Statistical analysis was performed with JMP Pro 16.1.0 statistical software. Comparisons of categorical and continuous variables were performed using the Chi-square test and Student's t-test or the Mann-Whitney U test, respectively. A P-value of <0.05 was considered significant. Cumulative overall survival (OS) and recurrence-free survival (RFS) rates were calculated using the Kaplan-Meier method, and differences between curves were evaluated using the log-rank test.

Results

JMJD6 expression in ICC specimens

Patients with ICC comprised 34 males and 17 females at an age range of 33-82 years (median=60). JMJD6 expression was analyzed in ICC specimens. Immunohistochemical analysis revealed that JMJD6 expression was predominantly noted in the nuclei (Fig. 2A and B). The percentage of stained tumor cells was analyzed, and the mean value was selected as the cut-off point to obtain comparable subgroup sizes.

Association of JMJD6 expression with clinicopathological features

The relationship between JMJD6 expression and clinicopathological factors in patients with ICC was evaluated (Table I). High JMJD6 expression was observed in 32 of 51 patients (62.7%). JMJD6 expression was not significantly associated with clinicopathological factors, except for older age in low-expression samples (65.6 vs. 58.7 years, P= 0.043).

Table I

Clinicopathological factors and JMJD6 expression in IHC.

Table I

Clinicopathological factors and JMJD6 expression in IHC.

Factors (Category)JMJD6 High expression (n=32)JMJD6 Low expression (n=19)P-value
Median age, years (IQR)58.7 (55-63)65.6 (60-71)0.043a
Sex  0.68
     Male (%)22(69)12(63) 
     Female (%)10(31)7(37) 
Albumin (g/dl)4.11 (3.97-4.27)4.03 (3.84-4.23)0.49
Total bilirubin (mg/dl)0.72 (0.31-1.13)1.31 (0.78-1.84)0.081
Tumor size (mm)44.9 (37.3-52.4)46.6 (37.3-55.9)0.77
Solitary/Multiple23/910/90.16
Microvascular invasion16/1413/50.20
Perihilar/Peripheral12/2010/90.29
Differentiation (well/mod/por)10/20/29/8/10.42
CEA (ng/ml)3.37 (1.88-4.86)3.12 (1.18-5.06)0.84
CA19-9 (U/ml)1349 (0-6,946)7,778 (863-14,693)0.15

[i] aP<0.05. CA19-9, carbohydrate antigen 19-9; CEA, carcinoembryonic antigen; JMJD6, Jumonji-domain containing 6.

JMJD6 expression and survival analysis

According to univariate analysis, low JMJD6 expression was significantly associated with poor overall survival (OS, P=0.0033) (Fig. 1C) and recurrence-free survival (RFS, P=0.048) (Fig. 1D).

Univariate analysis revealed that JMJD6 expression, carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9) were unfavorable predictors for OS. In the multivariate analysis, low JMJD6 expression and high CA19-9 were revealed to be the independent worse prognostic factors for OS (Table II). In the analyses regarding RFS, univariate analysis showed that low JMJD6 expression, low serum albumin level, high carcinoembryogenic antigen (CEA), and microvascular invasion were unfavorable predictors for RFS. In the multivariate analysis, low JMJD6 expression, low serum albumin level and high carcinoembryogenic antigen (CEA) were independent worse prognostic factors of RFS (Table III). The analyses were also performed in the subgroups of peripheral and perihilar ICC and did not reveal any specific differences (data not shown).

Table II

Univariate and multivariate Cox proportional hazard analyses of overall survival in patients with ICC. Bold numbers indicate statistically significant correlations (P<0.05).

Table II

Univariate and multivariate Cox proportional hazard analyses of overall survival in patients with ICC. Bold numbers indicate statistically significant correlations (P<0.05).

 Univariate analysisMultivariate analysis
FactorsHazard ratio (95% CI)P-valueHazard ratio (95% CI)P-value
JMJD6, low expression2.73 (1.36-5.48)0.0047a3.47 (1.62-7.45)0.0014a
Age ≥60 (year)1.20 (0.60-2.37)0.6070  
Sex, male1.39 (0.66-2.91)0.3890  
Albumin ≤3.5 (g/dl)1.87 (0.45-7.85)0.3917  
Total bilirubin ≥2.0 (mg/dl)2.99 (0.67-13.4)0.1522  
CEA ≥5 (U/ml)2.76 (1.18-6.46)0.0189a1.52 (0.61-3.81)0.3671
CA19-9 ≥50 (U/ml)2.08 (1.01-4.28)0.0461a2.72 (1.25-5.89)0.0114a
Tumor size ≥5-cm (cm)1.98 (0.99-3.97)0.0530  
MVI, positive1.94 (0.91-4.12)0.0845  
Location, perihilar1.26 (0.63-2.51)0.5108  
Number of tumors >11.95 (0.97-3.91)0.0593  

[i] aP<0.05. CA19-9, carbohydrate antigen 19-9; CEA, carcinoembryonic antigen; ICC, intrahepatic cholangiocarcinoma; JMJD6, Jumonji-domain containing 6; MVI, microvascular invasion.

Table III

Univariate and multivariate Cox proportional hazard analyses of recurrence-free survival in patients with ICC.

Table III

Univariate and multivariate Cox proportional hazard analyses of recurrence-free survival in patients with ICC.

 Univariate analysisMultivariate analysis
FactorsHazard ratio (95% CI)P-valueHazard ratio (95% CI)P-value
JMJD6, low expression2.20 (1.13-4.26)0.0206a2.33 (1.16-4.68)0.0179a
Age ≥60 (year)1.30 (0.67-2.50)0.4388  
Sex, male1.73 (0.83-3.60)0.1428  
Albumin ≤3.5 (g/dl)3.45 (1.03-11.5)0.0448a5.18 (1.45-18.5)0.0113a
Total bilirubin ≥2.0 (mg/dl)3.69 (0.84-16.2)0.0835  
CEA ≥5 (U/ml)4.10 (1.70-9.88)0.0016a3.80 (1.53-9.46)0.0041a
CA19-9 ≥50 (U/ml)1.58 (0.79-3.14)0.1973  
Tumor size ≥5 (cm)1.98 (0.99-3.97)0.0530  
MVI, positive2.32 (1.12-4.78)0.0231a1.97 (0.92-4.21)0.0804
Location, perihilar0.94 (0.55-2.06)0.8586  
Number of tumors >11.93 (0.97-3.81)0.0592  

[i] aP<0.05. CA19-9, carbohydrate antigen 19-9; CEA, carcinoembryonic antigen; ICC, intrahepatic cholangiocarcinoma; JMJD6, Jumonji-domain containing 6; MVI, microvascular invasion.

In vitro JMJD6 expression, cellular assays and RNA sequencing

To explore the importance of JMJD6 in ICC, JMJD6 was knocked down in SSP-25 cells. The migration assay (Fig. 2A) and the viability assay (Fig. 2B) did not show any significant difference between JMJD6-knockeddown cells and the control. RNA sequencing revealed 91 genes whose expression were positively altered and 167 negatively altered genes in JMJD6-depleted cells (Fig. 2C and D, Table SI). Among those, we focused on immunology-related genes to evaluate the impact of tumor immunology on the prognosis of ICC. Notably, JMJD6 knockdown increased the expression of PD-L1. An increase in PD-L1 expression under JMJD6 depletion was also identified using RT-PCR (Fig. 2E). Western blotting also revealed similar increase of PD-L1 expression in ICC cells after JMJD6 knockdown (Fig. 2F).

PD-L1 in clinical samples

To investigate the PD-L1 expression in clinical specimens, we performed IHC for PD-L1 in the same samples used for the IHC for JMJD6 (Fig. 3A). The result showed that 34 of 51 samples were positive for PD-L1. The rate of high PD-L1 expression was higher in the low JMJD6 group and vice versa (Fig. 3B; P=0.025). We also performed the survival analysis of PD-L1 expression, but it did not show any significant results (P-value for OS=0.3070, P-value for PFS=0.0687, data not shown).

Discussion

Patients with ICC have a poor prognosis, and curative treatments, such as surgical resection, are limited to early-stage disease. Systemic therapy options and their effectiveness are also limited. In contrast to the dramatic decrease of hepatocellular carcinoma owing to the development of virological treatment and newly emerged systemic therapy options, ICC treatment is limited and still requires further investigation (20).

In this study, we showed that the epigenetic regulator JMJD6 is a favorable prognostic factor for ICC and that JMJD6 is a possible regulator of PD-L1 expression. A mechanism that can explain these results is that JMJD6 modifies the promoter region of PD-L1 and inhibits PD-L1 expression. JMJD6 demethylates histones and other proteins, and it promotes or inhibits gene expression depending on which amino acid and on what site they are located. Also, PD-L1 expression is reported to be upregulated through epigenetic regulation with histone demethylases (21). Thus, JMJD also has the possibility to regulate the expression of PD-L1.

The relationship between JMJD6 and tumor has previously been reported as an unfavorable prognostic factor in various cancers, including breast cancer, colon cancer, oral squamous cancer, melanoma, and hepatocellular carcinoma. However, to the best of our knowledge, the role of JMJD6 in ICC has not been investigated (22-24).

Our results showed that JMJD6 is a good prognostic factor, in contrast to past reports. As JMJD6 is an epigenetic modifier, the influence of the protein varies depending on what type of protein to regulate, e.g., CDK4 in HCC, p53 in colon cancer. Therefore, we performed RNA sequencing to elucidate the genes regulated by JMJD6, which resulted in the identification of 171 candidate genes. Our institution has previously reported the impact of tumor immunology on cancer prognosis (12,25,26); thus, we focused on immune-related genes. We found that PD-L1 mRNA levels increased in response to JMJD6 knockdown, indicating that JMJD6 regulates PD-L1 expression. This inverse relationship between JMJD6 and PD-L1 expression was confirmed by PCR, western blotting, and IHC.

Although in vitro experiments have revealed the connection between JMJD6 and PD-L1, this was not sufficient to explain that PD-L1 expression is controlled by JMJD6, and the key to connect them is the epigenetic modification. CD274, the gene encoding PD-L1, is located on chromosome 9p24.1. In this region, genomic regulation has been proven to upregulate PD-L1 expression, resulting in immune escape (27). Among these types of regulation, H3K4me3 is upregulated by MLL1, an H3K4 methylation-specific histone methyltransferase in pancreatic cancer (28). The present data suggest that a similar mechanism is possibly activated to mediate PD-L1 expression.

PD-L1 induces cancer cell immune evasion by binding to the PD-1 receptor on activated T cells, which results in tolerance of tumor-reactive T cells, rendering tumor cells resistant to CD8+ T cells (29). In the current study, although the positivity of PD-L1 was not sufficient to demonstrate any correlation with the prognosis of ICC, the high JMJD6 expression, which inversely reflected PD-L1 expression, impacted the prognosis of ICC. Also, the use of immune checkpoint inhibitor combined with the conventional systemic therapy has emerged to be effective in prolonging the survival of biliary tract cancer patients. Thus, JMJD6 is a potential biomarker to prove the susceptibility of ICI in each individual.

This study has several limitations. First, the current study was derived from only one institution and the number of samples was small. Second, we did not perform any epigenetic experiments to directly prove the mechanism, by analysis with chromatin immunoprecipitation (ChIP) for example. Also, our cellular experiments only focused on viability and migration, and we did not perform the ones regarding apoptosis or cell cycle, which were reported in previous JMJD6 reports. Therefore, there is a room for future research about these factors. Additionally, the current research only presented the in vitro experiments and clinical sample study. Further studies using in vivo tumor mouse models and anti-PD-L1 agents are required to obtain more persuasive evidence.

In conclusion, JMJD6 is an independent favorable prognostic factor for ICC and is a candidate target protein for the treatment of ICC, focusing on the tumor microenvironment.

Supplementary Material

RNA sequenc ing results of altered gene expression in JMJD6 depleted cells

Acknowledgements

Not applicable.

Funding

Funding: This study was supported by the following grants: Grants-in-Aid (KAKENHI) from the Ministry of Health, Labour and Welfare, Japan (grant no. JP-19K09198).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YKF and SI carried out the molecular studies, participated in the sequence alignment and drafted the manuscript. KY carried out the evaluation of IHC. TF and DO performed the RNA sequencing. SI, TT and NH participated in the design of the study. YO, TY and MM conceived of the study and participated in its design and coordination and helped to draft the manuscript. YKF and SI confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

This study was approved by the Clinical Research Ethics Committee of Kyushu University Hospital according to the Ethical Guidelines of the Japanese Government (approval no. 30-578). The patients provided the written consent for the use of their tissues for future scientific research at the time of collection.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Zhang H, Yang T, Wu M and Shen F: Intrahepatic cholangiocarcinoma: Epidemiology, risk factors, diagnosis and surgical management. Cancer Lett. 379:198–205. 2016.PubMed/NCBI View Article : Google Scholar

2 

Doherty B, Nambudiri VE and Palmer WC: Update on the diagnosis and treatment of cholangiocarcinoma. Curr Gastroenterol Rep. 19(2)2017.PubMed/NCBI View Article : Google Scholar

3 

Blechacz B and Gores GJ: Cholangiocarcinoma: Advances in pathogenesis, diagnosis, and treatment. Hepatology. 48:308–321. 2008.PubMed/NCBI View Article : Google Scholar

4 

Yugawa K, Itoh S, Kurihara T, Yoshiya S, Mano Y, Takeishi K, Harada N, Ikegami T, Soejima Y, Mori M and Yoshizumi T: Skeletal muscle mass predicts the prognosis of patients with intrahepatic cholangiocarcinoma. Am J Surg. 218:952–958. 2019.PubMed/NCBI View Article : Google Scholar

5 

Sakamoto Y, Kokudo N, Matsuyama Y, Sakamoto M, Izumi N, Kadoya M, Kaneko S, Ku Y, Kudo M, Takayama T, et al: Proposal of a new staging system for intrahepatic cholangiocarcinoma: Analysis of surgical patients from a nationwide survey of the liver cancer study group of Japan. Cancer. 122:61–70. 2016.PubMed/NCBI View Article : Google Scholar

6 

Fadok VA, Bratton DL, Rose DM, Pearson A, Ezekewitz RA and Henson PM: . A receptor for phosphatidylserine-specific clearance of apoptotic cells. Nature. 405:85–90. 2000.PubMed/NCBI View Article : Google Scholar

7 

Chang B, Chen Y, Zhao Y and Bruick RK: JMJD6 is a histone arginine demethylase. Science. 318:444–447. 2007.PubMed/NCBI View Article : Google Scholar

8 

Unoki M, Masuda A, Dohmae N, Arita K, Yoshimatsu M, Iwai Y, Fukui Y, Ueda K, Hamamoto R, Shirakawa M, et al: Lysyl 5-hydroxylation, a novel histone modification, by Jumonji domain containing 6 (JMJD6). J Biol Chem. 288:6053–6062. 2013.PubMed/NCBI View Article : Google Scholar

9 

Yang J, Chen S, Yang Y, Ma X, Shao B, Yang S, Wei Y and Wei X: Jumonji domain-containing protein 6 protein and its role in cancer. Cell Prolif. 53(e12747)2020.PubMed/NCBI View Article : Google Scholar

10 

Nolz JC: Molecular mechanisms of CD8(+) T cell trafficking and localization. Cell Mol Life Sci. 72:2461–2473. 2015.PubMed/NCBI View Article : Google Scholar

11 

Farhood B, Najafi M and Mortezaee K: CD8+ cytotoxic T lymphocytes in cancer immunotherapy: A review. J Cell Physiol. 234:8509–8521. 2019.PubMed/NCBI View Article : Google Scholar

12 

Yugawa K, Itoh S, Yoshizumi T, Iseda N, Tomiyama T, Toshima T, Harada N, Kohashi K, Oda Y and Mori M: Prognostic impact of tumor microvessels in intrahepatic cholangiocarcinoma: Association with tumor-infiltrating lymphocytes. Mod Pathol. 34:798–807. 2021.PubMed/NCBI View Article : Google Scholar

13 

Asahi Y, Hatanaka KC, Hatanaka Y, Kamiyama T, Orimo T, Shimada S, Nagatsu A, Sakamoto Y, Kamachi H, Kobayashi N, et al: Prognostic impact of CD8+ T cell distribution and its association with the HLA class I expression in intrahepatic cholangiocarcinoma. Surg Today. 50:931–940. 2020.PubMed/NCBI View Article : Google Scholar

14 

Ronnekleiv-Kelly SM and Pawlik TM: Staging of intrahepatic cholangiocarcinoma. Hepatobiliary Surg Nutr. 6:35–43. 2017.PubMed/NCBI View Article : Google Scholar

15 

Fontugne J, Augustin J, Pujals A, Compagnon P, Rousseau B, Luciani A, Tournigand C, Cherqui D, Azoulay D, Pawlotsky JM and Calderaro J: PD-L1 expression in perihilar and intrahepatic cholangiocarcinoma. Oncotarget. 8:24644–24651. 2017.PubMed/NCBI View Article : Google Scholar

16 

Yugawa K, Itoh S, Yoshizumi T, Iseda N, Tomiyama T, Morinaga A, Toshima T, Harada N, Kohashi K, Oda Y and Mori M: CMTM6 stabilizes PD-L1 expression and is a new prognostic impact factor in hepatocellular carcinoma. Hepatol Commun. 5:334–348. 2020.PubMed/NCBI View Article : Google Scholar

17 

Berardo C, Siciliano V, Di Pasqua LG, Richelmi P, Vairetti M and Ferrigno A: Comparison between Lipofectamine RNAiMAX and GenMute transfection agents in two cellular models of human hepatoma. Eur J Histochem. 63(3048)2019.PubMed/NCBI View Article : Google Scholar

18 

Izumi T, Sakata K, Okuzaki D, Inokuchi S, Tamura T, Motooka D, Nakamura S, Ono C, Shimokawa M, Matsuura Y, et al: Characterization of human pegivirus infection in liver transplantation recipients. J Med Virol. 91:2093–2100. 2019.PubMed/NCBI View Article : Google Scholar

19 

Shimokawa M, Yoshizumi T, Itoh S, Iseda N, Sakata K, Yugawa K, Toshima T, Harada N, Ikegami T and Mori M: Modulation of Nqo1 activity intercepts anoikis resistance and reduces metastatic potential of hepatocellular carcinoma. Cancer Sci. 111:1228–1240. 2020.PubMed/NCBI View Article : Google Scholar

20 

Feng M, Pan Y, Kong R and Shu S: Therapy of primary liver cancer. Innovation (Camb). 1(100032)2020.PubMed/NCBI View Article : Google Scholar

21 

Sheng W, LaFleur MW, Nguyen TH, Chen S, Chakravarthy A, Conway JR, Li Y, Chen H, Yang H, Hsu PH, et al: LSD1 ablation stimulates anti-tumor immunity and enables checkpoint blockade. Cell. 174:549–563.e19. 2018.PubMed/NCBI View Article : Google Scholar

22 

Wan J, Liu H, Yang L, Ma L, Liu J and Ming L: JMJD6 promotes hepatocellular carcinoma carcinogenesis by targeting CDK4. Int J Cancer. 144:2489–2500. 2019.PubMed/NCBI View Article : Google Scholar

23 

Wong M, Sun Y, Xi Z, Milazzo G, Poulos RC, Bartenhagen C, Bell JL, Mayoh C, Ho N, Tee AE, et al: JMJD6 is a tumorigenic factor and therapeutic target in neuroblastoma. Nat Commun. 10(3319)2019.PubMed/NCBI View Article : Google Scholar

24 

Wang F, He L, Huangyang P, Liang J, Si W, Yan R, Han X, Liu S, Gui B, Li W, et al: JMJD6 promotes colon carcinogenesis through negative regulation of p53 by hydroxylation. PLoS Biol. 12(e1001819)2014.PubMed/NCBI View Article : Google Scholar

25 

Itoh S, Yoshizumi T, Yugawa K, Imai D, Yoshiya S, Takeishi K, Toshima T, Harada N, Ikegami T, Soejima Y, et al: Impact of immune response on outcomes in hepatocellular carcinoma: Association with vascular formation. Hepatology. 72:1987–1999. 2020.PubMed/NCBI View Article : Google Scholar

26 

Yugawa K, Itoh S, Iseda N, Kurihara T, Kitamura Y, Toshima T, Harada N, Kohashi K, Baba S, Ishigami K, et al: Obesity is a risk factor for intrahepatic cholangiocarcinoma progression associated with alterations of metabolic activity and immune status. Sci Rep. 11(5845)2021.PubMed/NCBI View Article : Google Scholar

27 

Ikeda S, Okamoto T, Okano S, Umemoto Y, Tagawa T, Morodomi Y, Kohno M, Shimamatsu S, Kitahara H, Suzuki Y, et al: PD-L1 is upregulated by simultaneous amplification of the PD-L1 and JAK2 genes in non-small cell lung cancer. J Thorac Oncol. 11:62–71. 2016.PubMed/NCBI View Article : Google Scholar

28 

Lu C, Paschall AV, Shi H, Savage N, Waller JL, Sabbatini ME, Oberlies NH, Pearce C and Liu K: The MLL1-H3K4me3 axis-mediated PD-L1 expression and pancreatic cancer immune evasion. J Natl Cancer Inst. 109(djw283)2017.PubMed/NCBI View Article : Google Scholar

29 

Gong J, Chehrazi-Raffle A, Reddi S and Salgia R: Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: A comprehensive review of registration trials and future considerations. J Immunother Cancer. 6(8)2018.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

August-2022
Volume 17 Issue 2

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kosai-Fujimoto Y, Itoh S, Yugawa K, Fukuhara T, Okuzaki D, Toshima T, Harada N, Oda Y, Yoshizumi T, Mori M, Mori M, et al: Impact of JMJD6 on intrahepatic cholangiocarcinoma. Mol Clin Oncol 17: 131, 2022
APA
Kosai-Fujimoto, Y., Itoh, S., Yugawa, K., Fukuhara, T., Okuzaki, D., Toshima, T. ... Mori, M. (2022). Impact of JMJD6 on intrahepatic cholangiocarcinoma. Molecular and Clinical Oncology, 17, 131. https://doi.org/10.3892/mco.2022.2564
MLA
Kosai-Fujimoto, Y., Itoh, S., Yugawa, K., Fukuhara, T., Okuzaki, D., Toshima, T., Harada, N., Oda, Y., Yoshizumi, T., Mori, M."Impact of JMJD6 on intrahepatic cholangiocarcinoma". Molecular and Clinical Oncology 17.2 (2022): 131.
Chicago
Kosai-Fujimoto, Y., Itoh, S., Yugawa, K., Fukuhara, T., Okuzaki, D., Toshima, T., Harada, N., Oda, Y., Yoshizumi, T., Mori, M."Impact of JMJD6 on intrahepatic cholangiocarcinoma". Molecular and Clinical Oncology 17, no. 2 (2022): 131. https://doi.org/10.3892/mco.2022.2564