Altered mRNA expression levels of the major components of sphingolipid metabolism, ceramide synthases and their clinical implication in colorectal cancer

  • Authors:
    • Sung Won Jang
    • Woo‑Jae Park
    • Hyeonji Min
    • Taeg Kyu Kwon
    • Seong Kyu Baek
    • Ilseon Hwang
    • Shin Kim
    • Jong‑Wook Park
  • View Affiliations

  • Published online on: September 18, 2018     https://doi.org/10.3892/or.2018.6712
  • Pages: 3489-3500
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ceramide synthases (CerSs) synthesize various ceramides of different acyl chain lengths and serve important roles in the proliferation and death of cancer cells by regulating sphingolipid metabolism‑related signaling pathways. The present study investigated the mRNA expression levels of various CerS genes using mRNA expression data from six independent colorectal cancer (CRC) cohorts and a Korean CRC dataset. Expression levels of CERS2, CERS5 and CERS6 mRNA were significantly increased in the majority of the studied groups. However, CERS4 expression was only significantly altered in two groups. Additionally, a positive correlation was observed between altered CERS4 and CERS5 mRNA levels in The Cancer Genome Atlas Colon and Rectal Cancer dataset. Notably, CERS2 and CERS4, as well as CERS5 and CERS6 levels, were positively correlated with each other in Korean patients with CRC. However, the mRNA expression levels of these four CerS genes were not associated with any clinicopathological characteristics in Korean patients with CRC. Finally, overexpressing CERS2 or CERS6 inhibited the in vitro viability of various CRC cells. Taken together, these findings indicated that CERS2, CERS4, CERS5, and CERS6 are significantly dysregulated in CRC, suggesting they may serve important roles in the pathophysiology of this malignancy.

Introduction

Colorectal cancer (CRC) is a malignancy derived from the colorectal epithelium and is the third most commonly diagnosed cancer type worldwide (1). Although the mortality rates of CRC have been decreasing due to screening, reduced risk factor prevalence and/or improved therapies (2,3), CRC remains a global health burden in terms of morbidity and mortality, with ~700,000 estimated mortalities annually (1). It has been reported that the complicated and complex pathogenetic mechanisms of CRC involve genomic rearrangements, chromatin remodeling, genetic mutations and epigenetic changes (4,5).

The sphingolipid rheostat is a proposed concept that may regulate cell fate decisions (6). The two major components of the sphingolipid rheostat are ceramide and sphinogosine-1 phosphate, which are interconvertible sphingolipid metabolites that regulate cell growth and survival by modulating sphingolipid rheostat-related signaling (6,7). Ceramide has tumor suppressive anticancer properties, including potentiating signaling networks that drive apoptosis, autophagy and cell cycle arrest (8). Ceramide synthases (CerSs) are integral membrane proteins of the endoplasmic reticulum that synthesize ceramides of different acyl chain lengths. To date, six CerS families have been identified in mammals (9). Dysregulation of CerS activity has been reported to be associated with tumor cell invasion (10), proliferation (11), apoptosis (12) and epithelial-mesenchymal transition (13), as well as with the prognosis of patients with cancer (14). For example, in head and neck squamous cell carcinoma, downregulation of CERS1 leads to apoptotic resistance (15), while CERS1 overexpression enhances growth-inhibitory effects (16). Additionally, CERS2, CERS4 and CERS6 mRNA expression levels are increased in breast cancer (17), and the upregulation of CERS4 and CERS6 leads to reduced cell proliferation and the induction of apoptosis (18). Given these results and the association of altered CerS expression with malignant transformation, the present study aimed to characterize the mRNA expression of various CerS genes in CRC and non-neoplastic adjacent tissues (NST).

The present study investigated the mRNA expression levels of various CerS genes using mRNA expression data from six independent CRC cohorts and a Korean CRC dataset. Furthermore, the clinical significance of altered CerS genes expression was evaluated in the Korean CRC dataset.

Materials and methods

Gene expression databases and cluster analysis

Gene expression RNAseq dataset (Level 3) and clinical data for The Cancer Genome Atlas Colon and Rectal Cancer (TCGA-COADREAD) cohort (19) were downloaded from the UCSC Xena (https://xena.ucsc.edu). CRC gene expression microarray data used in this study were downloaded from the publicly available GEO databases (http://www.ncbi.nlm.nih.gov/geo/): GSE21815 (20), GSE44076 (21), GSE44861 (22), GSE41258 (23) and GSE33113 (24). The GEO datasets used in this study include 562 CRC tissues and 222 NST from respective same patient groups. The downloaded raw data of GEO databases were normalized at the transcript and gene level using the Robust Multichip Average method (25). Cluster analysis was performed using Cluster 3.0 to classify the samples into statistically similar groups, and the resulting heatmaps were visualized in TreeView 1.6 (www.eisenlab.org/eisen). The four CerS genes present in the TCGA COADREAD, GSE44076 and GSE44861 cohorts were LASS2, LASS4, LASS5 and LASS6. The present study meets the publication guidelines provided by TCGA.

Patients and tissues

A total of 59 patients (mean age, 64.83±9.48; age range, 38–83; 34 males and 25 females) diagnosed with CRC were included in the present study. CRC and NSTs were obtained from patients undergoing surgery in Keimyung University Dongsan Medical Center (Daegu, Korea) between April 2008 and January 2010. Enrolled patients with CRC were classified according to the AJCC Tumor-Node Metastasis (TNM) staging criteria (26). Tissue samples were immediately frozen in liquid nitrogen and stored at −196°C until RNA isolation. Tissue samples were provided by Keimyung Human Bio-Resource Bank (Daegu, Korea). Written informed consent was obtained from each study participant and the protocols were approved by the Institutional Review Board of Keimyung University Dongsan Medical Center (approval no. 2015-11-059-001).

RNA isolation and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total cellular RNA was extracted from tissues using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA). RNA was quantified using NanoDrop 1000 (Thermo Fisher Scientific, Inc.). Each cDNA was synthesized from 2 µg total RNA using MMLV reverse transcriptase (Promega Corporation, Madison, WI, USA), according to the manufacturer's protocol. qPCR was performed on the LightCycler® 480 Real-Time PCR system (Roche Diagnostics GmbH, Mannheim, Germany) using the specific primer pairs presented in Table I and SYBR-Green Premix (Toyobo Life Science, Osaka, Japan). The qPCR was performed using the following thermocycling conditions: 95°C for 10 min; followed by 45 cycles of 95°C for 10 sec, 60°C for 10 sec, and 72°C for 12 sec. Melting curve was analyzed to determine primer specificity. b-actin was used as a housekeeping gene for normalization, and a no-template sample was used as a negative control. qPCR data were analyzed using the 2−∆∆Cq method (27). Each experiment was performed three times.

Table I.

Primer sequences used in quantitative polymerase chain reaction.

Table I.

Primer sequences used in quantitative polymerase chain reaction.

PrimerSequence
CERS2 sense 5′-ATCGTCTTCGCCATTGTTTT-3′
CERS2 antisense 5′-GGCAGGATAGAGCTCCAGTG-3′
CERS4 sense 5′-GGAGGCCTGTAAGATGGTCA-3′
CERS4 antisense 5′-GAGGACCAGTCGGGTGTAGA-3′
CERS5 sense 5′-TGGAATTGGCCTTCTATTGG-3′
CERS5 antisense 5′-CAATGGTGACCAAGTGATGC-3′
CERS6 sense 5′-TGCCATTCTGGAAAAGGTCT-3′
CERS6 antisense 5′-ATGCTTCGAACATCCCAGTC-3′
β-actin sense 5′-CAGCCATGTACGTTGCTATCCAGG-3′
β-actin antisense 5′-AGGTCCAGACGCAGGATGGCATG-3′

[i] CERS, ceramide synthase mRNA.

Statistical analysis

Statistical analysis was performed using SPSS 22.0 (IBM Corp., Armonk, NY, USA). The cell viability data were analyzed using one-way analysis of variance and the Student-Newman-Keuls post hoc test. Differences between the groups were analyzed statistically using Student's t-test or Mann Whitney U test. The co-expression of the mRNAs of various CerS genes in TCGA-COADREAD cohort were searched using cBioPortal (http://cbioportal.org) (28). The association between inter-individual mRNA expression levels of CerS genes in Korean patients with CRC was assessed using Pearson's correlation coefficient analysis for continuous variables. Clinicopathological associations with the mRNA expression levels of various CerS genes in Korean CRC were analyzed using the Linear by linear association, the Pearson's Chi-square test and the Fisher's exact test for categorical variables. The mean value was used as the cut-off value (low and high) for categorical variables. P<0.05 was considered to indicate a statistically significant difference.

Transient transfection

Various human colorectal adenocarcinoma cell lines, HCT116, HT29, SW403 and SW480 cells, were plated onto 6-well plates at a density 7×105 cells/well and cultured overnight. pcDNA3.1-empty vector was used for plasmid constructs, including HA-tagged form of CERS2 (HA-CERS2) and HA-tagged form of CERS6 (HA-CERS6) constructs. All plasmids, including pcDNA3.1-empty vector, HA-CERS2 and HA-CERS6 were provided by Professor Anthony H. Futerman (Weizmann Institute of Science, Rehovot, Israel). The CRC cells were transfected with pcDNA3.1-empty vector, 2 µg HA-CERS2 and HA-CERS6 plasmid in 6-well plates using Lipofectamine reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. At 24 h after plasmid transfection, the subsequent experiments were conducted.

Western blot analysis

The transient transfected CRC cells were collected and washed twice with cold PBS, and cell pellets were prepared by suspending in modified radioimmunoprecipitation assay buffer (50 mM Tris-HCl pH 7.4, 1% NP-40, 0.25% Na-deoxycholate, 150 mM NaCl, 1 mM Na3VO4 and 1 mM NaF) containing protease inhibitors (100 µM phenylmethylsulfonyl fluoride, 10 µg/ml leupeptin, 10 µg/ml pepstatin and 2 mM EDTA). The lysates were centrifuged at 10,000 × g for 10 min at 4°C, and the supernatant fractions were collected. The total protein concentration was measured using Micro BCA™ Protein assay kit (Thermo Fisher Scientific, Inc.), according to the manufacturers protocol. Cellular proteins (60 mg) were mixed with protein 5X sample buffer (Elpis Biotech., Inc., Daejeon, Korea) and heated at 95°C for 5 min. The proteins were separated by 10% SDS-PAGE and then electrotransferred to Immobilon-P membranes (EMD Millipore, Billerica, MA, USA). The membranes were then blocked at room temperature with 5% skimmed dried milk in PBS/0.1% Tween-20 for 1 h, and incubated overnight at 4°C with anti-HA (1:2,000; mouse monoclonal; cat. no. SAB1411737) and anti-β-actin (1:2,000; mouse monoclonal; cat. no. A5441; both Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). The membranes were then washed six times with PBS/0.1% Tween-20 (30 min each) and incubated with the corresponding secondary antibodies (horseradish peroxidase-conjugated, horse antibodies to mouse IgG; 1:2,000; cat. no. 7076; Cell Signaling Technology, Inc.) for 1 h at room temperature. Following washing six times in PBS/0.1% Tween-20, the specific protein bands were detected using an enhanced chemiluminescence western blotting kit (EMD Millipore), according to the manufacturer's protocol.

Results

Altered expression levels of sphingolipid metabolism-related genes in six independent CRC cohorts

To investigate whether the sphingolipid metabolism-related genes (29) are dysregulated in CRC tissues, the present study re-analyzed the raw data of six independent CRC cohorts. To begin with, the cancer gene expression RNAseq datasets of 380 CRC patients were taken from the TCGA-COADREAD cohort through UCSC Xena. Next, CRC gene expression microarray data were downloaded from the publicly available Gene Expression Omnibus databases. The CRC gene expression microarrays, GSE21815, GSE33113, GSE41258, GSE44076 and GSE44861, were analyzed for potential transcriptome changes. Hierarchical clustering revealed that various sphingolipid metabolism-related genes were dysregulated in carcinomatous tissues compared with NST of patients with CRC (Fig. 1). To identify the significance of altered mRNA expression levels between CRC and NST, Student's t-test or Mann Whitney U test were performed (P<0.05). As demonstrated in Fig. 2, hierarchical clustering revealed that various sphingolipid metabolism-related genes were significantly dysregulated in CRC tissues compared with NST from the same patient groups. The list of analyzed sphingolipid metabolism-related genes is presented in Table II. Sphingosine kinase 1 (SPHK1) and UDP-glucose glycoprotein glucosyltransferase 2 (UGGT2) were significantly upregulated in the CRC tissues of all cohorts, while 15-hydroxyprostaglandin dehydrogenase (HPGD), lysophosphatidic acid receptor 1 (LPAR1), N-acylethanolamine acid amidase (NAAA), sphingomyelin phosphodiesterase 1 (SMPD1) and sphingomyelin phosphodiesterase acid-like 3A (SMPDL3A) were significantly downregulated in the CRC tissues of all cohorts (Fig. 2 and Table II).

Table II.

List of the analyzed genes involved in sphingolipid metabolism (Student's t-test, Mann Whitney U test; P<0.05).

Table II.

List of the analyzed genes involved in sphingolipid metabolism (Student's t-test, Mann Whitney U test; P<0.05).

Dataset TCGA-COADREADa GSE21815b GSE44076a GSE44861a GSE41258a GSE33113b
No. analyzed genes362035192415
Gene symbolA4GALTB4GALT6ASAH1ASAHL (NAAA)ASAH1CERS5
ASAH1BECN1B4GALT6B4GALT6BECN1CERS6
B4GALNT1CERKBECN1BNIP3CERKHPGD
B4GALT6CERS2CERKCERKCERS2LPAR1
BECN1CERS5DEGS1DEGS1CERS6NAAA
CERKDEGS1GAL3ST1EDG2 (LPAR1)GAL3ST1NSMAF
GAL3ST1GBAGBAEDG5 (S1PR2)HPGDSFTPB
GALCHPGDHPGDHPGDLCTSMPD1
GBALPAR1LASS1CERS2LPAR1SMPDL3A
HPGDNAAALASS2NSMAFLPAR2SMPDL3B
LASS1S1PR4LASS5SMPD1NAAASPHK1
LASS2SLC26A10LASS6SMPD2NSMAFSPTLC1
LASS3SMPD1LCTSMPDL3AS1PR1SPTLC2
LASS4SMPDL3ALPAR1SMPDL3BSLC26A10ST6GALNAC5
LASS5SMPDL3BLPAR2SPHK1SMPD1UGGT2
LASS6SPHK1NAAASPHK2SMPDL3A
LPAR1ST3GAL5NSMAFSPTLC1SMPDL3B
LPAR2ST8SIA1S1PR1ST6GALNAC5SPHK1
NAAAUGGT1S1PR4UGCGL2 (UGGT2)SPHK2
NSMAFUGGT2SFTPB ST3GAL5
S1PR1 SLC26A10 ST8SIA1
S1PR4 SMPD1 UGCG
SLC26A10 SMPD2 UGGT1
SMPD1 SMPDL3A UGGT2
SMPDL3A SMPDL3B
SMPLL3B SPHK1
SPHK1 SPHK2
SPHK2 SPTLC1
SPTLC2 SPTLC2
ST3GAL5 ST3GAL5
ST6GALNAC5 ST8SIA1
ST8SIA1 ST8SIA3
ST8SIA3 UGCG
UGCG UGGT1
UGGT1 UGGT2
UGGT2

a Student's t-test

b Mann Whitney U test. TCGA-COADREAD, The Cancer Genome Atlas Colon and Rectal Cancer.

Dysregulation of various CerSs in six independent CRC cohorts

Next, the present study evaluated whether the mRNA expression levels of the four CerS genes, which are abundant in colorectal tissues (30), are dysregulated in human CRC specimens with respect to NST. As demonstrated in Fig. 3, among six cohorts, CERS2 mRNA levels were significantly increased in five independent cohorts, while CERS5 and CERS6 were significantly upregulated in four independent cohorts. The specific platforms of each cohort and their associated studies are listed in Table III.

Table III.

mRNA expression levels of CerS gene in colorectal cancer tissues of patients from various datasets used in the present study.

Table III.

mRNA expression levels of CerS gene in colorectal cancer tissues of patients from various datasets used in the present study.

DatasetPlatformCERS2CERS4CERS5CERS6(Refs.)
TCGA (COADREAD)RNA sequencingUpDownUpUp(19)
GSE21815Human Whole Genome Microarray 4×4K G4112FUpN/AUpN/A(20)
GSE44076Affymetrix Human Genome U219 ArrayUpN/AUpUp(21)
GSE44861Affymetrix HT Human Genome U133A ArrayUpN/AN/AN/A(22)
GSE41258Affymetrix U133A ArrayUpN/AN/AUp(23)
GSE33113Affymetrix Human Genome U133 Plus 2.0 ArrayN/AN/AUpUp(24)

[i] CERS, ceramide synthase; TCGA-COADREAD, The Cancer Genome Atlas Colon and Rectal Cancer.

Altered CerS genes mRNA expression in Korean patients with CRC

To determine whether there is altered CERS2, CERS4, CERS5 and/or CERS6 mRNA expression in Korean patients with CRC, the expression levels of these four CerSs were measured using qPCR in 59 paired CRC and NST specimens from Korean patients. Following exclusion of unqualified results, the qPCR data were analyzed. The present study revealed that mRNA expression levels of all four CerS genes were significantly upregulated in CRC tissues compared with corresponding NSTs (CERS2, P<0.001; CERS4, P=0.006; CERS5, P<0.001; CERS6, P<0.001; Fig. 4; Table IV).

Table IV.

mRNA expression levels of CerS gene in CRC tissues as compared with NST of Korean patients with CRC.

Table IV.

mRNA expression levels of CerS gene in CRC tissues as compared with NST of Korean patients with CRC.

CERS family CERS2 CERS4 CERS5 CERS6
No. patients 59 55 49 55
Type of tissueNST CRCNST CRCNST CRCNST CRC
Mean (ΔCq value)5.85 4.265.68 4.438.89 7.857.26 4.51
Regulation Up Up Up Up
P-value <0.001 0.06 <0.001 <0.001

[i] Student t-test. CERS, ceramide synthase; NST, non-neoplastic surrounding colon tissues; CRC, colorectal cancer.

Exogenous CERS2 and CERS6 expression decreases the viability of human CRC cells

It has previously been observed that CERS6-overexpression reduces the proliferation of CRC cells and induces apoptosis, whereas CERS2-overexpression increases the proliferation of CRC cells (18). To confirm the effect of overexpressing CerSs in CRC cells, HCT116, HT29, SW403 and SW480 cells were transiently transfected with constructs to overexpress HA-CERS2 and HA-CERS6, respectively. After 48 and 72 h, the numbers of viable cells were counted using a hemocytometer. As demonstrated in Fig. 5, overexpression of CERS2 and CERS6 decreased the viability of this panel of CRC cell lines.

Inter-individual associations between mRNA expression levels of CerS genes in patients with CRC

Combinational patterns of CerS gene expression, including CerS hetero-complexes and co-expression of CerS genes, serve important roles in sphingolipid metabolism (31,32). Therefore, associations between the mRNA levels of each CerS gene were identified in the TCGA-COADREAD cohort and in the Korean CRC cohort. Using cBioPortal to analyze the TCGA-COADREAD cohort, co-expression analysis revealed that CERS4 and CERS5 had high correlation coefficients (Pearson's correlation=0.36; Spearman's correlation=0.48; Fig. 6A). Next, these correlations were assessed using Pearson's correlation coefficient analysis in the 59 Korean patients with CRC. There were significant correlations between CERS2 and CERS4, and also between CERS5 and CERS6, with a Pearson's correlation coefficient value of 0.532 (P<0.001; Fig. 6B) and 0.439 (P=0.003; Fig. 6C), respectively. Furthermore, significant correlations between the mRNA expression levels of CERS2 and CERS4 (P=0.009) and of CERS5 and CERS6 (P<0.001) were identified using Fisher's exact test (Table V).

Table V.

Association between mRNA expression levels of various CerS genes and clinicopathological parameters in Korean patients with colorectal cancer.

Table V.

Association between mRNA expression levels of various CerS genes and clinicopathological parameters in Korean patients with colorectal cancer.

CERS2 expressionCERS4 expressionCERS5 expressionCERS6 expression




ParameterLowHighP-valueLowHighP-valueLowHighP-valueLowHighP-value
Sex 1.000b 0.195b 0.719b 1.000b
  Male244 1711 208 226
  Female142 133 133 133
Age, years 0.606b 1.000b 0.408b 0.659b
  ≤5082 7 391 91
  >50304 2311 2410 268
T stage 0.609c 0.675c 0.457c 0.140c
  T120 20 02 02
  T271 44 80 71
  T3244 217 208 235
  T451 33 51 51
N stage 0.063c 0.061c 0.055c 0.288c
  N0252 216 189 216
  N171 53 62 53
  N263 45 90 90
  N300 00 00 00
M stage 0.456b 0.581b 0.558b 0.566b
  Negative355 2812 2911 319
  Positive31 22 40 40
BMI 0.653c 0.320c 0.593c 0.566c
  ≤18.500 00 00 00
  18.5–24.910 10 10 10
  25-29.9274 1912 229 256
  >30102 102 102 93
CEA 1.000b 0.540a 0.706b 0.703b
  >5112 85 94 112
  ≤5274 229 247 247
CERS2 0.009b 0.630b 1.000b
  Low 299 299 308
  High 15 42 51
CERS4 0.009b 0.456b 0.233b
  Low291 219 228
  High95 122 131
CERS5 0.630b 0.456b <0.001b
  Low294 2112 312
  High92 92 47
CERS6 1.000b 0.233b <0.001b
  Low305 2213 314
  High81 81 27

a Pearson's χ2 test

b Fisher's exact test

c Linear by linear association. CERS, ceramide synthase; BMI, body mass index; CEA, carcinoembryonic antigen.

Association between mRNA expression levels of CerS genes and clinicopathological parameters of Korean patients with CRC

To determine the clinicopathological implications of dysregulated expression of specific CerS genes in CRC, the association between CerS gene mRNA level and clinicopathological characteristics, which are used to represent progression and aggressiveness, were evaluated. Prior to the statistical analysis, the 44 patients, whose clinical data were available, were classified according to each clinicopathological characteristic (Table V). The results obtained from the statistical analysis of the Korean cohort revealed that altered mRNA expression levels of CerS genes were not significantly associated with any clinical parameters, including sex, age, Tumor-Node-Metastasis stage, body mass index or carcinoembryonic antigen titer.

Discussion

Sphingolipid metabolism serves a critical role in mammalian cell growth arrest and survival (33). Accumulating evidence have demonstrated that CerS, a major component in sphingolipid metabolism (7), regulates various biological phenomenon, including apoptosis (34), cancer (17,35), ER stress (36), hepatopathy (37), hypoxia/re-oxygenation injury (38), lipid metabolism (39), neurodegeneration (40), and sensitivity to chemotherapeutic drugs and radiation (30). Although aberrant CerS expression is correlated with cell death and proliferation (1014) in various types of cancer, much uncertainty remains regarding the dysregulated mRNA levels of CerS gene in CRC and the clinical implications of this.

The aims of the present study were to investigate the mRNA expression levels and functions of CerS genes, which are primarily expressed in the intestine (30,41), and analyze their clinicopathological implications in patients with CRC. To begin with, significantly dysregulated sphingolipid metabolism-related genes were identified in the heat-maps of 6 independent CRC cohorts (Fig. 2). The hierarchical clustering results demonstrated considerable dysregulation of sphingolipid metabolism-related genes in CRC tissues compared with corresponding NST of independent CRC cohorts. Among the considerably altered genes, certain genes were overlapping over 6 independent cohorts. SPHK1 and UGGT2 were significantly upregulated in CRC tissues. This result is in accordance with those of recent studies that indicated that SPHK1 is overexpressed and serves an important role in tumorigenesis, proliferation, invasiveness and metastasis in CRC (42,43). On the other hand, HPGD, LPAR1, NAAA, SMPD1 and SMPDL3A were all significantly downregulated in CRC tissues. HPGD is a cytoplasmic enzyme responsible for degrading PGE2 in colorectal tissue (44), and functions as a tumor suppressor gene in various types of cancer (4548). The present study observed that HPGD was downregulated in 6 independent CRC cohorts (Fig. 2). However, little is known regarding the cellular functions and clinicopathological implications of LPAR1, NAAA, SMPD1, SMPDL3A and UGGT2 in CRC. Therefore, further studies investigating the functional role of these genes in CRC are required as these transcripts may be diagnostic markers or promising therapeutic candidates.

Additionally, the differential mRNA levels of CERS2, CERS4, CERS5 and CERS6 in CRC and NST were analyzed in 1,001 patients with CRC from 6 independent publicly-available CRC cohorts and a cohort of Korean patients with CRC. The results of the present study should be interpreted with caution as qPCR, RNA-Seq and microarray are different experimental platforms with different sensitivities, principles and dynamic ranges. Nonetheless, the results revealed that CERS2 was significantly upregulated in the majority of cohorts (Fig. 3; Table III) and in the cohort of Korean patients with CRC (Fig. 4; Table IV). It was recently demonstrated that CERS2-overexpression had no effect on the viability of HCT116 cells, whereas overexpressing CERS2 plus the addition of very-long chain acyl-CoAs significantly enhanced colony formation in HCT116 cells (18). Unlike these previous results, the present study revealed that CERS2-overexpression reduced the viability of various human CRC cells, including HCT116 cells (Fig. 5). The primary technical differences between these two experiments are the culture time following transfection and the use of different expression plasmids. Additionally, knockdown experiments were performed using shRNAs against CERS2 mRNA and CERS6 mRNA, knockdown of CerS2 or CerS6 did not affect the proliferation of CRC SW403 and SW480 cells (data not shown). Although the exact mechanism that underlies the effect on cell viability was not elucidated in the present study, it is possible that sustained cell culture time following transfection may affect the synthesis of ceramides of various chain lengths.

It has been reported that increased expression of CerS6 and C16:0-Ceramide resulted in a sensitization of SW620 cells to TRAIL-induced apoptosis (49), and CERS6-overexpression significantly inhibited the colony formation capacity and increased the apoptosis of HCT116 cells (18). In accordance with these previous results, the present study demonstrated that CERS6 is significantly upregulated in CRC tissues, compared with NST (Figs. 3 and 4; Table IV) and CERS6-overexpression led to inhibition of cell viability in various human CRC cells (Fig. 5). Notably, controversial results have demonstrated that CerS6 and C16:0-Ceramide protected cells against ER-stress in human head and neck squamous cell carcinomas (36). Although oncogenes are usually upregulated in cancer tissues compared with non-neoplastic tissues, previous studies and the results presented in the present study indicated that the roles of CerSs and ceramides of specific chain lengths are complicated and cell type-dependent. Notably, it was demonstrated that CERS4 was significantly upregulated, but only in the cohort of Korean patients with CRC (Fig. 4), while it was downregulated in the TCGA-COADREAD cohort (Fig. 3). Future studies specifically focused on CERS4 in different CRC populations are required in order to understand this phenomena.

Additionally, the present study evaluated correlations between inter-individual mRNA expression levels of CerS genes and their clinicopathological implications in patients with CRC. A recent study revealed that inter-individual differences in the mRNA expression levels of CerS genes are significantly correlated with each other in cancer tissues (17). Furthermore, Combinational patterns of CerS expression are involved in sphingolipid metabolism (31,32). The results of analyzing correlations between inter-individual CerS genes mRNA expression levels revealed a correlation between CERS4 and CERS5 in TCGA-COADREAD, between CERS2 and CERS4, and between CERS5 and CERS6 in the cohort of Korean patients with CRC. However, combinational patterns of CerS expression may be associated with sphingolipid metabolism. Therefore, it will be important to determine which components serve critical roles in sphingolipid metabolism in different disease and tissue settings. To the best of our knowledge, the present study was the first to investigate the clinicopathological implications of dysregulated CerS genes mRNA expression in CRC. However, no correlation was observed between mRNA expression levels of specific CerS genes and the investigated clinicopathological parameters.

In conclusion, the present study revealed that the mRNA expression levels of CERS2, CERS4, CERS5 and CERS6 were significantly upregulated or downregulated in various independent CRC cohorts, suggesting that dysregulated CerS gene expression may serve a role in CRC development.

Acknowledgements

The authors would like to thank all members of their research group for providing enthusiastic participation in the present study. The biospecimens for the present study were provided by the Keimyung Human Bio-Resource Bank, a member of the National Biobank of Korea, which is supported by the Ministry of Health and Welfare. All samples derived from the National Biobank of Korea were obtained following receipt of written informed consent and Institutional Review Board approval.

Funding

The present study was supported by the National Research Foundation of Korea Grant funded by the Korean Government (Ministry of Science, ICT & Future Planning; grant nos. 2017R1C1B5016670 and 2014R1A5A2010008).

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request. The primary and processed data used to generate the analyses presented here can be downloaded by registered users from The Cancer Genome Atlas at http://tcga-data.nci.nih.gov/tcga/tcgaDownload.jsp.

Authors' contributions

SWJ, WJP and SK contributed to the conception and design of the study, analysis of the data, interpretation of results and the writing of the manuscript. SWJ, WJP, HM and SK contributed to the acquisition of data. SWJ, WJP, HM, SKB, IH and SK performed the experiments. TKK contributed to the conception and design of the study. JWP and IH contributed to the conception and design of the study and provided guidance regarding the clinical implications of the study. WJP and SK reviewed and edited the manuscript. All authors read and approved the manuscript, and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

The experimental study was approved by the Institutional Review Board of Keimyung University Dongsan Medical Center (approval no. 2015-11-059-001). Written informed consent was obtained from each study participant.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interest.

Glossary

Abbreviations

Abbreviations:

CRC

colorectal cancer

CerS

ceramide synthase

TCGA-COADREAD

The Cancer Genome Atlas Colon and Rectal Cancer

qPCR

quantitative polymerase chain reaction

NST

non-neoplastic surrounding colon tissues

LPAR1

lysophosphatidic acid receptor 1

NAAA

N-acylethanolamine acid amidase

SPHK1

sphingosine kinase 1

HPGD

15-hydroxyprostaglandin dehydrogenase

SMPD1

sphingomyelin phosphodiesterase 1

SMPDL3A

sphingomyelin phosphodiesterase acid-like 3A

UGGT2

UDP-glucose glycoprotein glucosyltransferase 2

HA-CERS2

HA-tagged form of CERS2

HA-CERS6

HA-tagged form of CERS6

References

1 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Edwards BK, Ward E, Kohler BA, Eheman C, Zauber AG, Anderson RN, Jemal A, Schymura MJ, Lansdorp-Vogelaar I, Seeff LC, et al: Annual report to the nation on the status of cancer, 1975–2006, featuring colorectal cancer trends and impact of interventions (risk factors, screening, and treatment) to reduce future rates. Cancer. 116:544–573. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Bosetti C, Levi F, Rosato V, Bertuccio P, Lucchini F, Negri E and La Vecchia C: Recent trends in colorectal cancer mortality in Europe. Int J Cancer. 129:180–191. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Fearon ER and Vogelstein B: A genetic model for colorectal tumorigenesis. Cell. 61:759–767. 1990. View Article : Google Scholar : PubMed/NCBI

5 

Fearon ER: Molecular genetics of colorectal cancer. Ann Rev Pathol. 6:479–507. 2011. View Article : Google Scholar

6 

Cuvillier O, Pirianov G, Kleuser B, Vanek PG, Coso OA, Gutkind S and Spiegel S: Suppression of ceramide-mediated programmed cell death by sphingosine-1-phosphate. Nature. 381:800–803. 1996. View Article : Google Scholar : PubMed/NCBI

7 

Newton J, Lima S, Maceyka M and Spiegel S: Revisiting the sphingolipid rheostat: Evolving concepts in cancer therapy. Exp Cell Res. 333:195–200. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Morad SA and Cabot MC: Ceramide-orchestrated signalling in cancer cells. Nat Rev Cancer. 13:51–65. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Stiban J, Tidhar R and Futerman AH: Ceramide synthases: Roles in cell physiology and signaling. Adv Exp Med Biol. 688:60–71. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Fan SH, Wang YY, Lu J, Zheng YL, Wu DM, Zhang ZF, Shan Q, Hu B, Li MQ and Cheng W: CERS2 suppresses tumor cell invasion and is associated with decreased V-ATPase and MMP-2/MMP-9 activities in breast cancer. J Cell Biochem. 116:502–513. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Chen J, Li X, Ma D, Liu T, Tian P and Wu C: Ceramide synthase-4 orchestrates the cell proliferation and tumor growth of liver cancer in vitro and in vivo through the nuclear factor-κB signaling pathway. Oncol Lett. 14:1477–1483. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Suzuki M, Cao K, Kato S, Komizu Y, Mizutani N, Tanaka K, Arima C, Tai MC, Yanagisawa K, Togawa N, et al: Targeting ceramide synthase 6-dependent metastasis-prone phenotype in lung cancer cells. J Clin Invest. 126:254–265. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Edmond V, Dufour F, Poiroux G, Shoji K, Malleter M, Fouqué A, Tauzin S, Rimokh R, Sergent O, Penna A, et al: Downregulation of ceramide synthase-6 during epithelial-to-mesenchymal transition reduces plasma membrane fluidity and cancer cell motility. Oncogene. 34:996–1005. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Fitzgerald S, Sheehan KM, Espina V, O'Grady A, Cummins R, Kenny D, Liotta L, O'Kennedy R, Kay EW and Kijanka GS: High CerS5 expression levels associate with reduced patient survival and transition from apoptotic to autophagy signalling pathways in colorectal cancer. J Pathol Clin Res. 1:54–65. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Separovic D, Breen P, Joseph N, Bielawski J, Pierce JS, VAN Buren E and Gudz TI: siRNA-mediated down-regulation of ceramide synthase 1 leads to apoptotic resistance in human head and neck squamous carcinoma cells after photodynamic therapy. Anticancer Res. 32:2479–2485. 2012.PubMed/NCBI

16 

Senkal CE, Ponnusamy S, Rossi MJ, Bialewski J, Sinha D, Jiang JC, Jazwinski SM, Hannun YA and Ogretmen B: Role of human longevity assurance gene 1 and C18-ceramide in chemotherapy-induced cell death in human head and neck squamous cell carcinomas. Mol Cancer Ther. 6:712–722. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Erez-Roman R, Pienik R and Futerman AH: Increased ceramide synthase 2 and 6 mRNA levels in breast cancer tissues and correlation with sphingosine kinase expression. Biochem Biophys Res Commun. 391:219–223. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Hartmann D, Lucks J, Fuchs S, Schiffmann S, Schreiber Y, Ferreirós N, Merkens J, Marschalek R, Geisslinger G and Grösch S: Long chain ceramides and very long chain ceramides have opposite effects on human breast and colon cancer cell growth. Int J Biochem Cell Biol. 44:620–628. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Cancer Genome Atlas Network: Comprehensive molecular characterization of human colon and rectal cancer. Nature. 487:330–337. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Kogo R, Shimamura T, Mimori K, Kawahara K, Imoto S, Sudo T, Tanaka F, Shibata K, Suzuki A, Komune S, et al: Long noncoding RNA HOTAIR regulates polycomb-dependent chromatin modification and is associated with poor prognosis in colorectal cancers. Cancer Res. 71:6320–6326. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Sanz-Pamplona R, Berenguer A, Cordero D, Molleví DG, Crous-Bou M, Sole X, Paré-Brunet L, Guino E, Salazar R, Santos C, et al: Aberrant gene expression in mucosa adjacent to tumor reveals a molecular crosstalk in colon cancer. Mol Cancer. 13:462014. View Article : Google Scholar : PubMed/NCBI

22 

Ryan BM, Zanetti KA, Robles AI, Schetter AJ, Goodman J, Hayes RB, Huang WY, Gunter MJ, Yeager M, Burdette L, et al: Germline variation in NCF4, an innate immunity gene, is associated with an increased risk of colorectal cancer. Int J Cancer. 134:1399–1407. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Sheffer M, Bacolod MD, Zuk O, Giardina SF, Pincas H, Barany F, Paty PB, Gerald WL, Notterman DA and Domany E: Association of survival and disease progression with chromosomal instability: A genomic exploration of colorectal cancer. Proc Natl Acad Sci USA. 106:7131–7136. 2009. View Article : Google Scholar : PubMed/NCBI

24 

de Sousa E, Melo F, Colak S, Buikhuisen J, Koster J, Cameron K, de Jong JH, Tuynman JB, Prasetyanti PR, Fessler E, van den Bergh SP, et al: Methylation of cancer-stem-cell-associated Wnt target genes predicts poor prognosis in colorectal cancer patients. Cell Stem Cell. 9:476–485. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U and Speed TP: Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 4:249–264. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Edge SB and Cancer AJCo: AJCC Cancer Staging Handbook: Form the AJCC Cancer Staging Manual. Springer; New York: 2010

27 

Schmittgen TD and Livak KJ: Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 3:1101–1108. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al: The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2:401–404. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Ruckhäberle E, Rody A, Engels K, Gaetje R, von Minckwitz G, Schiffmann S, Grösch S, Geisslinger G, Holtrich U, Karn T, et al: Microarray analysis of altered sphingolipid metabolism reveals prognostic significance of sphingosine kinase 1 in breast cancer. Breast Cancer Res Treat. 112:41–52. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Mullen TD, Hannun YA and Obeid LM: Ceramide synthases at the centre of sphingolipid metabolism and biology. Biochem J. 441:789–802. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Park JW, Park WJ and Futerman AH: Ceramide synthases as potential targets for therapeutic intervention in human diseases. Biochim Biophys Acta. 1841:671–681. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Mesicek J, Lee H, Feldman T, Jiang X, Skobeleva A, Berdyshev EV, Haimovitz-Friedman A, Fuks Z and Kolesnick R: Ceramide synthases 2, 5, and 6 confer distinct roles in radiation-induced apoptosis in HeLa cells. Cell Signal. 22:1300–1307. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Mandala SM, Thornton R, Tu Z, Kurtz MB, Nickels J, Broach J, Menzeleev R and Spiegel S: Sphingoid base 1-phosphate phosphatase: A key regulator of sphingolipid metabolism and stress response. Proc Natl Acad Sci USA. 95:150–155. 1998. View Article : Google Scholar : PubMed/NCBI

34 

Panjarian S, Kozhaya L, Arayssi S, Yehia M, Bielawski J, Bielawska A, Usta J, Hannun YA, Obeid LM and Dbaibo GS: De novo N-palmitoylsphingosine synthesis is the major biochemical mechanism of ceramide accumulation following p53 up-regulation. Prostaglandins Other Lipid Mediat. 86:41–48. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Karahatay S, Thomas K, Koybasi S, Senkal CE, Elojeimy S, Liu X, Bielawski J, Day TA, Gillespie MB, Sinha D, et al: Clinical relevance of ceramide metabolism in the pathogenesis of human head and neck squamous cell carcinoma (HNSCC): Attenuation of C18-ceramide in HNSCC tumors correlates with lymphovascular invasion and nodal metastasis. Cancer Lett. 256:101–111. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Senkal CE, Ponnusamy S, Bielawski J, Hannun YA and Ogretmen B: Antiapoptotic roles of ceramide-synthase-6-generated C16-ceramide via selective regulation of the ATF6/CHOP arm of ER-stress-response pathways. FASEB J. 24:296–308. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Pewzner-Jung Y, Brenner O, Braun S, Laviad EL, Ben-Dor S, Feldmesser E, Horn-Saban S, Amann-Zalcenstein D, Raanan C, Berkutzki T, et al: A critical role for ceramide synthase 2 in liver homeostasis: II. insights into molecular changes leading to hepatopathy. J Biol Chem. 285:10911–10923. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Jin J, Hou Q, Mullen TD, Zeidan YH, Bielawski J, Kraveka JM, Bielawska A, Obeid LM, Hannun YA and Hsu YT: Ceramide generated by sphingomyelin hydrolysis and the salvage pathway is involved in hypoxia/reoxygenation-induced Bax redistribution to mitochondria in NT-2 cells. J Biol Chem. 283:26509–26517. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Pewzner-Jung Y, Park H, Laviad EL, Silva LC, Lahiri S, Stiban J, Erez-Roman R, Brügger B, Sachsenheimer T, Wieland F, et al: A critical role for ceramide synthase 2 in liver homeostasis: I. alterations in lipid metabolic pathways. J Biol Chem. 285:10902–10910. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Spassieva SD, Ji X, Liu Y, Gable K, Bielawski J, Dunn TM, Bieberich E and Zhao L: Ectopic expression of ceramide synthase 2 in neurons suppresses neurodegeneration induced by ceramide synthase 1 deficiency. Proc Natl Acad Sci USA. 113:5928–5933. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Laviad EL, Albee L, Pankova-Kholmyansky I, Epstein S, Park H, Merrill AH Jr and Futerman AH: Characterization of ceramide synthase 2: Tissue distribution, substrate specificity, and inhibition by sphingosine 1-phosphate. J Biol Chem. 283:5677–5684. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Bao Y, Guo Y, Zhang C, Fan F and Yang W: Sphingosine kinase 1 and sphingosine-1-phosphate signaling in colorectal cancer. Int J Mol Sci. 18:E21092017. View Article : Google Scholar : PubMed/NCBI

43 

Long J, Xie Y, Yin J, Lu W and Fang S: SphK1 promotes tumor cell migration and invasion in colorectal cancer. Tumour Biol. 37:6831–6836. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Smartt HJ, Greenhough A, Ordóñez-Morán P, Talero E, Cherry CA, Wallam CA, Parry L, Al Kharusi M, Roberts HR, Mariadason JM, et al: β-catenin represses expression of the tumour suppressor 15-prostaglandin dehydrogenase in the normal intestinal epithelium and colorectal tumour cells. Gut. 61:1306–1314. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Wolf I, O'Kelly J, Rubinek T, Tong M, Nguyen A, Lin BT, Tai HH, Karlan BY and Koeffler HP: 15-hydroxyprostaglandin dehydrogenase is a tumor suppressor of human breast cancer. Cancer Res. 66:7818–7823. 2006. View Article : Google Scholar : PubMed/NCBI

46 

Ding Y, Tong M, Liu S, Moscow JA and Tai HH: NAD+-linked 15-hydroxyprostaglandin dehydrogenase (15-PGDH) behaves as a tumor suppressor in lung cancer. Carcinogenesis. 26:65–72. 2005. View Article : Google Scholar : PubMed/NCBI

47 

Song HJ, Myung SJ, Kim IW, Jeong JY, Park YS, Lee SM, Nam WH, Ryu YM, Fink SP, Yang DH, et al: 15-hydroxyprostaglandin dehydrogenase is downregulated and exhibits tumor suppressor activity in gastric cancer. Cancer Invest. 29:257–265. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Myung SJ, Rerko RM, Yan M, Platzer P, Guda K, Dotson A, Lawrence E, Dannenberg AJ, Lovgren AK, Luo G, et al: 15-Hydroxyprostaglandin dehydrogenase is an in vivo suppressor of colon tumorigenesis. Proc Natl Acad Sci USA. 103:12098–12102. 2006. View Article : Google Scholar : PubMed/NCBI

49 

White-Gilbertson S, Mullen T, Senkal C, Lu P, Ogretmen B, Obeid L and Voelkel-Johnson C: Ceramide synthase 6 modulates TRAIL sensitivity and nuclear translocation of active caspase-3 in colon cancer cells. Oncogene. 28:1132–1141. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2018
Volume 40 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jang SW, Park WJ, Min H, Kwon TK, Baek SK, Hwang I, Kim S and Park JW: Altered mRNA expression levels of the major components of sphingolipid metabolism, ceramide synthases and their clinical implication in colorectal cancer. Oncol Rep 40: 3489-3500, 2018
APA
Jang, S.W., Park, W., Min, H., Kwon, T.K., Baek, S.K., Hwang, I. ... Park, J. (2018). Altered mRNA expression levels of the major components of sphingolipid metabolism, ceramide synthases and their clinical implication in colorectal cancer. Oncology Reports, 40, 3489-3500. https://doi.org/10.3892/or.2018.6712
MLA
Jang, S. W., Park, W., Min, H., Kwon, T. K., Baek, S. K., Hwang, I., Kim, S., Park, J."Altered mRNA expression levels of the major components of sphingolipid metabolism, ceramide synthases and their clinical implication in colorectal cancer". Oncology Reports 40.6 (2018): 3489-3500.
Chicago
Jang, S. W., Park, W., Min, H., Kwon, T. K., Baek, S. K., Hwang, I., Kim, S., Park, J."Altered mRNA expression levels of the major components of sphingolipid metabolism, ceramide synthases and their clinical implication in colorectal cancer". Oncology Reports 40, no. 6 (2018): 3489-3500. https://doi.org/10.3892/or.2018.6712