2023 Impact Factor
Pancreatic cancer is the seventh leading cause of cancer-related deaths worldwide, climbing to the fourth in developed countries (Sung
Numerous studies have sought to identify cancer biomarkers employing omics approaches, focusing on diagnosis, prognosis, and chemotherapy resistance (Hristova and Chan, 2019; Cheong
Taurine, or 2-aminoethanesulfonic acid, is a semi-essential, non-proteinogenic amino acid and is one of the most abundant amino acids in eukaryotes, constituting up to 0.1% of human body weight (Ripps and Shen, 2012). Taurine plays pivotal roles in various biological processes, including bile acid conjugation, cardiovascular protection, central nervous system function, and anti-oxidation (Lourenco and Camilo, 2002). A recent study showed that taurine concentrations in humans inversely correlated with age-related diseases such as hypertension, inflammation, and type 2 diabetes, suggesting a potential link between taurine deficiency and aging (Singh
ADO, or 2-aminoethanethiol dioxygenase, is a thiol dioxygenase in humans that adds two oxygen atoms to cysteamine, forming hypotaurine (Dominy
In this study, we investigated the PDAC-specific metabolic pathways by applying multi-omics on PDAC tissues and explored their association with patients’ clinical phenotypes.
Pancreatic tissues were collected at Asan Medical Center (Seoul, Korea) from 2011 to 2020. A total of 108 samples (54 pairs of each tumor and matched normal tissues) were collected from patients diagnosed with PDAC. Written informed consent had been obtained from the patients. The study protocol was approved by the institutional review board (IRB No.: 2019-0185). Tissue samples were snap-frozen in liquid nitrogen, transferred to cryotubes and stored at −80°C. Tissue samples were transported to the measurement laboratory in boxes filled with dry ice and stored at −80°C until metabolomics analysis.
For metabolite extraction, 40 mg of tissue samples were weighed, snap-frozen in liquid nitrogen, and pulverized. Samples were lysed in a 600 μL of methanol/chloroform (2:1) followed by three cycles of vortexing, freezing in liquid nitrogen, and thawing at room temperature. After adding 400 μL of chloroform/water (1:1) mixture, samples were centrifuged at 15,000×g, 4°C for 20 min. The upper phase containing metabolites was separated, dried in a vacuum evaporator, and redissolved in 100 μL of acetonitrile/water (1:1) for analysis, with same volume of samples pooled for quality control (QC) samples. For the metabolite extraction from cells, incubated cells were lysed and processed following our previously reported protocol (Oh
For untargeted liquid chromatography-high resolution mass spectrometry (LC-HRMS) analysis, a Q Exactive Plus Hybrid Quadrupole-Orbitrap Mass Spectrometer coupled with Vanquish UHPLC (Thermo Scientific, Waltham, MA, USA) was used. Chromatic separation occurred on a BEH amide column (Waters 186004801, Milford, MA, USA) under column at 40°C and autosampler at 4°C. The QC sample was injected every ten samples. Two microliters of analytes were injected and eluted with a mobile phase of 10 mM ammonium acetate in water (A, pH 10.0) and 80% acetonitrile (B, pH 10.0). Gradient conditions were as follows: 0-2 min 100% B, 2-7.5 min gradient 100-60% B, 7.5-16 min 60% B, 16.5-20 min gradient 60-100% B, under flow rate of 0.2 mL/min at 0-16.5 min, and 0.3 mL/min at 16.5-20 min. Both positive and negative modes for tissue samples, and only positive mode for cell extracts were used, detecting mass range of 70-900.
LC-HRMS data were analyzed using MZmine 2.53, where chromatograms were generated and peaks isolated, identifying compounds via PubChem and the Human Metabolome Database. The sum of peak intensities from four features in ESI(+) mode was calculated as the intensity of taurine in tissue samples. The taurine peak from cell samples was detected using Thermo Xcalibur software (Thrermo Scientific, Waltham, MA, USA) with base peak m/z=122.022 and normalized against the total chromatogram peak sum.
Two human tissue microarrays (PA1001c and PA2072a, US Biomax, Derwood, MD, USA) with normal pancreas (45 cores, 19 cases) and PDAC (258 cores, 99 cases) were used to assess taurine levels, with tumor stage, metastasis and grade details provided by the manufacturer. For immunostaining, 8-μm sections underwent deparaffinization, antigen retrieval in citrate buffer (pH 6.0), peroxidase quenching with 3% H2O2 in phosphate-buffered saline (PBS), water rinse, and blocking with goat serum. Primary antibody incubation was done at 4°C for two days using a 1:30 anti-taurine antibody (AB5022, Sigma-Aldrich, St. Louis, MO, USA), followed by biotinylated secondary antibodies (1:60, BA-1000, Vector Laboratories, Newark, CA, USA) for 2 h at room temperature. Sections were then washed, treated with avidin-biotin complex, developed with 3,3′-diaminobenzidine, and counterstained with hematoxylin. One human tissue microarray (PA1001c, US Biomax) with normal pancreas (20 cores, 10 cases) and PDAC (78 cores, 39 cases) was used to examine the expression level of ADO. Immunostaining for ADO was performed in same protocol as taurine with modified antibody reaction, with an anti-ADO antibody (1:50, sc-515318, Santa Cruz, Dallas, TX, USA) and a 1:100 dilution of secondary antibody. All staining was scored by four levels (0 for no, 1 for weak, 2 for moderate, and 3 for strong staining).
The gene expression and the patient’s survival information were from the UCSC Xena website (The Cancer Genome Atlas [TCGA] TARGET The Genotype-Tissue Expression [GTEx] cohort, https://xenabrowser.net). In PDAC, tumor tissues (n=179) were all from TCGA, and among the corresponding normal tissues (n=171), 4 were from TCGA and the others were from GTEx. Gene expression profiles across various cancer types were analyzed through the GEPIA web server (http://gepia.cancer-pku.cn) (Tang
PANC-1, a human pancreatic cancer cell line, was purchased from Korean Cell Line Bank. Cells were grown in DMEM (LM 011-01, Welgene, Gyeongsan, Korea) supplemented with 10% fetal bovine serum and 1% penicillin-streptomycin, at 37°C incubator. Three siRNAs (control, ADO-KD1, and ADO-KD2) were generated by Bioneer (Daejeon, Korea), and the sequences for ADO silencing followed those from a previous study (Shen
Total RNA was extracted using Easy-Spin Total RNA Extraction Kit (iNtRON Biotechnology, Seongnam, Korea), and cDNA was synthesized with the High-Capacity cDNA Reverse Transcription Kit (Applied Biosystems, Waltham, MA, USA), all according to the manufacturer’s protocols. The primer sequences are listed in Supplementary Table 2. qPCR was performed using TOPreal SyBR Green qPCR Premix (Enzynomics, Daejeon, Korea) on an Applied Biosystems Prism 7300 system. Western blot experiment was conducted using primary antibodies against ADO (16479-1-AP, Proteintech, Rosemont, IL, USA), β-actin (sc-47778, Santa Cruz), p65 (sc-8008, Santa Cruz), phospho-p65 (#3033, CST, Danvers, MA, USA), MEK1 (#9124, CST), phospho-MEK1 (Thr286) (#9127, CST), phospho-MEK1/2 (Ser221) (#2338, CST), ERK1/2 (#9102, CST), and phospho-ERK1/2 (#4377, CST).
Transfected PANC-1 cell (1.0×104 cells per well) were seeded into 96-well plates and incubated for 6 h for cell adhesion. After the cells were adhered, 10 µL of CCK solution (DonginLS, Seoul, Korea) was added to each well, followed by 2 h incubation. The measurement of absorbance at 450 nm was conducted daily for six days. For the clonogenic assay, 100 cells were seeded into 6-well plate. The cells were cultured for 20 days in DMEM supplemented with 10% fetal bovine serum and 1% penicillin-streptomycin. Once colonies had formed, the plate was washed with PBS and stained with 0.5% crystal violet solution for 30 min. Following staining, the plate was gently rinsed and left to dry before imaging was conducted.
Six-week-old female BALB/c mice were procured from Orient Bio (Gapyeong, Korea). All animal procedures were conducted following institutional guidelines and received approval from the Seoul National University Institutional Animal Care and Use Committee (Approval No. 2006241-2E). Prior to experimentation, the mice underwent a two-week acclimatization period. For tumor induction, PANC-1 cells, transfected with either control siRNA or ADO-KD1, were suspended in PBS and combined with an equal volume of Matrigel (Corning 354248, Corning, NY, USA). Two hundred microliters of mixture containing 5.0×106 cells were injected subcutaneously into the right flank of the mice. Following a month post-injection, body weight and tumor growth were monitored every ten days. Tumor volume (in mm3) was calculated using the formula: [length (mm)×width (mm)×width (mm)]/2.
Descriptive statistical analyses, encompassing paired, Student’s, and Welch’s t-test, Mann-Whitney U test, and receiver operating characteristic (ROC) analysis, were executed using GraphPad Prism 9.3 (GraphPad, Boston, MA, USA). Kaplan-Meier analysis was conducted in the Kaplan-Meier Plotter (https://kmplot.com). All statistical tests were two-sided, and
The demographic and clinical characteristics of the patients in this study are presented in Table 1. Fifty-four pairs of normal pancreas and tumor tissues from patients with PDAC were obtained during surgery. Of these 54 patients, 38 and 16 were diagnosed as stage I-II and III-IV, respectively [stage IA (n=4), IB (n=14), IIA (n=6), IIB (n=14), III (n=11), and IV (n=5)]. Classification of tumor stages was according to the 8th edition of the American Joint Committee on Cancer (Chun
Table 1 Demographic and clinical characteristics of patients (n=54)
Characteristics | n (%) | |
---|---|---|
Gender | Male | 30 (55.6) |
Female | 24 (44.4) | |
Age | <60 | 24 (44.4) |
≥60 | 30 (55.6) | |
Pre-operational CA19-9 | <35 U/mL | 18 (33.3) |
≥35 U/mL | 36 (66.7) | |
AJCC stage | IA | 4 (7.4) |
IB | 14 (25.9) | |
IIA | 6 (11.1) | |
IIB | 14 (25.9) | |
III | 11 (20.4) | |
IV | 5 (9.3) | |
Tumor grade | 1 (Well differentiated) | 5 (9.3) |
2 (Moderately differentiated) | 35 (64.8) | |
3 (Poorly differentiated) | 6 (11.1) | |
4 (Undifferentiated) | 1 (1.9) | |
N/A | 7 (13.0) | |
Lymphovascular invasion | No | 21 (38.9) |
Yes | 32 (59.3) | |
N/A | 1 (1.9) | |
Perineural invasion | No | 12 (22.2) |
Yes | 42 (77.8) | |
Adjuvant chemotherapy | No | 15 (27.8) |
Yes | 39 (72.2) | |
Recurrence | No or censored | 16 (29.6) |
Yes | 38 (70.4) |
CA19-9, carbohydrate antigen 19-9; AJCC, American Joint Committee on Cancer; N/A, not available.
We performed untargeted metabolomics analysis on 54 pairs of normal pancreas and pancreatic tumor samples using liquid chromatography-high resolution mass spectrometry (LC-HRMS). Comprehensive analysis of tissue metabolic profiles was conducted, followed by multivariate statistical analysis to discern differences between normal and tumor samples. The unsupervised multivariate principal component analysis (PCA) score plot demonstrated close clustering of quality control (QC) samples, indicating instrumental stability (Fig. 1A). An orthogonal projections to latent structure-discriminant analysis (OPLS-DA) model was obtained using one predictive (Pp) and two orthogonal components (Po) to discriminate the normal and PDAC samples and to identify the relevant biomarkers. The OPLS-DA score plot showed a good separation between normal and tumor groups with good predictive values (R2=0.737, Q2= 0.543; Fig. 1B). Among 296 features detected from both ESI(+) and ESI(−) modes of LC-HRMS, 109 features exhibited a significant decrease (
To validate the elevated taurine level in an independent group, we used pancreatic tissue microarrays that include both normal pancreas (47 cores, 19 cases) and PDAC tissues (258 cores, 99 cases) obtained commercially. In addition, we employed an orthogonal modality for measuring taurine level (IHC using antibody) to avoid a potential bias from the metabolomics approach. The IHC staining showed a significant elevation of taurine level in the PDAC tissues (
To identify the gene responsible for the elevation of taurine in PDAC, we first conducted bioinformatics screening focused on genes associated with the taurine metabolism pathway. Utilizing the KEGG (Kanehisa and Goto, 2000) (Kyoto Encyclopedia of Genes and Genomes) pathway database, 16 genes implicated in taurine metabolism were identified. Additionally, a taurine transporter gene (
Then, we analyzed the clinical relevance of
On the IHC analysis of independent tissue microarray comprising normal pancreas (20 cores, 10 cases) and PDAC tissues (78 cores, 39 cases). The IHC staining revealed a significant increase in ADO protein levels in PDAC tissues (
As the above data suggests association of the ADO-Taurine metabolic pathway with PDAC, we tested its causal involvement using genetic manipulation. For that, two distinct siRNAs targeting
To see if the ADO-Taurine’s causal involvement in PDAC cell growth is also manifested
In this study, we identified an elevated level of taurine in PDAC and established its relationship with cancer recurrence. Importantly, we showed the causal involvement of taurine metabolism in PDAC tumorigenesis through the
Interestingly, our bioinformatics analysis revealed that ADO mRNA levels are reduced in testicular germ cell tumors (TGCT) compared to normal testicular tissue, contrasting with our findings in PDAC. This difference highlights the tissue-specific roles of ADO and taurine metabolism in cancer. Taurine plays a crucial role in normal testicular function, being essential for sperm physiology and fertility (Li
In conclusion, our study underscores the pivotal role of the ADO-Taurine axis in PDAC’s altered metabolism, highlighting its significance in the disease’s progression and potential as a prognostic marker. Our findings, derived from multi-omics analyses and validated through
This work was supported by the National Research Foundation (NRF) grant funded by the Korean government (NRF-2021R1A5A2031612 to S-S.H.), a grant (2021IP0020-1 to S.C.K.) from Asan Institute for Life Sciences, Asan Medical Center, and a Inha University Grant (to K.H.J).
The authors declare that they have no conflict of interest.
Hoonsik Nam and Woohyung Lee contributed equally to this study.
Conception and design: S.C.K. and S.P. Development of methodology: H.N., W.L., S-S.H., S.C.K., and S.P. Clinical sample procurement: W.L. and S.C.K. Acquisition of data: H.N., W.L., Y.J.L., J-M.K., and K.H.J. Analysis and interpretation of data: H.N., W.L., Y.J.L., J-M.K., and K.H.J. Writing, review, and/or revision of manuscript: All authors. All authors read and approved the final manuscript.