Open Access

Causal association between metabolites and upper gastrointestinal tumors: A Mendelian randomization study

  • Authors:
    • Pengkhun Nov
    • Chongyang Zheng
    • Duanyu Wang
    • Syphanna Sou
    • Socheat Touch
    • Samnang Kouy
    • Peizan Ni
    • Qianzi Kou
    • Ying Li
    • Arzoo Prasai
    • Wen Fu
    • Kunpeng Du
    • Jiqiang Li
  • View Affiliations

  • Published online on: September 24, 2024     https://doi.org/10.3892/mmr.2024.13336
  • Article Number: 212
  • Copyright: © Nov 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

Upper gastrointestinal (UGI) tumors, notably gastric cancer (GC) and esophageal cancer (EC), are significant global health concerns due to their high morbidity and mortality rates. However, only a limited number of metabolites have been identified as biomarkers for these cancers. To explore the association between metabolites and UGI tumors, the present study conducted a comprehensive two‑sample Mendelian randomization (MR) analysis using publicly available genetic data. In the present study, the causal relationships were examined between 1,400 metabolites and UGI cancer using methods such as inverse variance weighting and weighted medians, along with sensitivity analyses for heterogeneity and pleiotropy. Functional experiments were conducted to validate the MR results. The analysis identified 57 metabolites associated with EC and 58 with GC. Key metabolites included fructosyllysine [EC: Odds ratio (OR)=1.450, 95% confidence interval (CI)=1.087‑1.934, P=0.011; GC: OR=1.728, 95% CI=1.202‑2.483, P=0.003], 2'‑deoxyuridine to cytidine ratio (EC: OR=1.464, 95% CI=1.111‑1.929, P=0.007; GC: OR=1.464, 95% CI=1.094‑1.957, P=0.010) and carnitine to protonylcarnitine (C3) ratio (EC: OR=0.655, 95% CI=0.499‑0.861, P=0.002; GC: OR=0.664, 95% CI=0.486‑0.906, P=0.010). Notably, fructosyllysine levels and the 2'‑deoxyuridine to cytidine ratio were identified as risk factors for both EC and GC, while the C3 ratio served as a protective factor. Functional experiments demonstrated that fructosyllysine and the 2'‑deoxyuridine to cytidine ratio promoted the proliferation of EC and GC cells, whereas carnitine inhibited their proliferation. In conclusion, the present findings provide insights into the causal factors and biomarkers associated with UGI tumors, which may be instrumental in guiding targeted dietary and pharmacological interventions, thereby contributing to the prevention and treatment of UGI cancer.

Introduction

Upper gastrointestinal (UGI) tumors, characterized by high morbidity and mortality rates, present significant global health challenges, especially esophageal cancer (EC; 510,716 new cases in 2022) and gastric cancer (GC; ~1 million new cases in 2022) (1). The prognosis for patients with UGI cancer remains poor in multiple countries, primarily because of insufficient screening initiatives (2,3). Elucidating the mechanisms that initiate and advance UGI cancers is essential to develop successful prevention and therapeutic approaches. Growing evidence shows that metabolites, small molecules intermediately produced during multiple cellular metabolic reactions, are implicated in the pathways leading to UGI tumors. Progress in molecular biology and the introduction of diverse omics techniques have markedly advanced epidemiological studies at the molecular level in this field (4).

Metabolic imbalance is increasingly recognized as a pivotal factor in the development of UGI tumors (5). Beyond changes in glucose metabolism, exemplified by the well-documented Warburg effect, disrupted metabolism has been reported in nucleotides, lipids and amino acids in both laboratory and clinical studies (68). Metabolites are a group of end products arising from complex interactions between inherent metabolism, genetic predispositions and environmental influences. High-throughput metabolomics enables comprehensive identification and quantification of a vast array of low molecular weight metabolites (<1,000 Da) within a single sample. This method is instrumental in identifying novel biomarkers and providing insights into the mechanisms of cancer causation (9,10). In addition, high-throughput metabolomics facilitates the discovery of new preventive measures and therapeutic targets (11). Previous research has analyzed a wide range of metabolites in UGI tumors using human samples such as urine, plasma and tissue (12,13). Although substantial efforts have been made in the field of UGI cancer metabolomics (14,15), these studies are predominantly descriptive. With the increase in research over recent years, there is a critical need for a comprehensive analysis to enhance our understanding of metabolomic profiles in UGI cancer, aiming to identify specific metabolites and consistently involved pathways.

Numerous cross-sectional, cohort, or retrospective studies have examined the association between metabolites and UGI tumors. However, due to their observational nature, these studies were restricted to identifying the correlations rather than the causations (16,17). While randomized controlled trials (RCTs), could potentially establish causality, interventions designed to manipulate metabolites are generally neither feasible nor ethical, thus limiting their ability to determine causal relationships. Given the constraints of both observational and interventional studies, Mendelian randomization (MR) in human genetics offers a powerful tool for rigorously investigating potential causal associations between elevated metabolite levels and UGI tumors (16,17). Traditional observational epidemiological approaches are susceptible to biases, rendering the association results from these studies prone to confounding factors (such as sex and age) and reverse causality (such as lifestyle changes due to UGI cancer), which lead to unreliable causal inferences (18). MR has become a primary genetic epidemiological research method, using genetic variations such as single nucleotide polymorphisms (SNPs) as instrumental variables to examine exposure factors and conclude causal relationships between exposures and outcomes. Due to the principle of randomly distributing alleles to offspring, the causal association estimates obtained from MR studies are not influenced by confounding factors. Additionally, since genes are determined before birth and cannot be altered by diseases, MR research effectively controls for the effects of reverse causality (19).

In the present study, MR was employed to explore the roles of metabolites in both histophysiological and pathophysiological processes leading to UGI tumors, using insights from a recent statistical analysis based on metabolite-focused Genome-Wide Association Study (GWAS) data (20). The present study aimed to investigate the causal association between 1,400 metabolites and UGI cancer, particularly focusing on their roles in tumor initiation, progression and treatment resistance. Moreover, functional experiments were conducted to further validate the findings from the MR analysis. The present study sought to provide insights that could enhance future metabolomic methodologies and advance etiological research, thereby supporting the development of precise prevention and innovative therapeutic strategies. It is anticipated that the findings will contribute to the creation of personalized treatment plans that target the specific metabolic vulnerabilities of UGI tumors.

Materials and methods

Study design

The cause-and-effect association between 1,400 metabolites and UGI tumors was assessed using two-sample MR analyses. MR utilized genetic variations as proxies for metabolic risk factors. To ensure reliable causal inference, the instrumental variables (IVs) used in MR were required to satisfy three critical assumptions: i) Genetic variation must be directly associated with the exposure (metabolites); ii) genetic variant should not be linked to any confounders that might affect the exposure and the outcome; and iii) the genetic variation should affect the outcome solely through the exposure, without any alternative pathways involved. The present study excluded points with P<1×105 from the outcome when conducting MR analysis to satisfy the second assumption. In addition, it used methodologies such as MR Egger (21) and MR-pleiotropy residual sum and outlier (PRESSO) to test and found no pleiotropy in the results for the third assumption. Furthermore, before conducting MR analysis, the present study had already searched for the corresponding SNPs on the GWAS catalog and removed the sites with pleiotropy. Although a number of measures had been taken to avoid environmental and genetic factors, it was hypothesized that the external environment can still have an influence. Finally, an experiment was performed to validate the present results (Fig. 1).

Data sources for exposure and outcome

The statistical summary of GWAS data for each metabolite was taken from the European GWAS website (http://www.ebi.ac.uk/gwas/; accession no. GCST90199621-90201020) (20). Cancer-specific keywords were applied to search relevant data for each cancer type (https://gwas.mrcieu.ac.uk/). Specifically, ukb-saige-150 was selected for EC identification, while ukb-saige-151 was used for GC screening. The UK Biobank, used for this data retrieval, is a substantial biomedical database and research resource aimed at enabling the exploration of genetic, environmental and lifestyle factors influencing various diseases and health outcomes (22). This database includes health and genetic information from >500,000 participants in the UK, making it one of the most comprehensive biomedical resources of its type.

GWAS studies aim to identify common genetic variants linked to complex disorders, ultimately guiding the development of translational prevention and treatment strategies. Given their extensive coverage of common SNPs and relative cost-effectiveness, GWAS has emerged as a valuable tool for clinical and commercial genetic testing. Essentially, GWAS acts as a potent tool that has greatly enhanced our knowledge of the genetic foundations of complex traits and diseases, establishing a robust foundation for future research and medical innovations. GWAS studies typically involve collecting and analyzing DNA samples from individuals to identify genetic variations associated with particular traits or diseases. The biological material collected for GWAS usually comprises DNA extracted from blood, saliva, or other tissues from participants. Researchers extract DNA from these samples and employ genotyping techniques to detect genetic variations, such as SNPs that might be linked to the trait or disease of interest. In the current study, data were obtained from the UK Biobank (www.ukbiobank.ac.uk) using specific ICD-10 (International Classification of Diseases 10th revision; accession nos. ukb-saige-150), and (GC: ukb-saige-151) IDs for each type of cancer. This involved analyzing the association between 1,400 types of metabolites and UGI tumors. Specifically, data included 394,092 European individuals (720 case patients and 393,372 control participants) for EC and 393,926 European individuals (554 case patients and 475,308 control participants) for GC. The association between metabolites and each cancer type was subsequently analyzed according to these IDs.

Instrument selection

Considering the extensive number of SNPs demonstrating genome-wide significance (P<5×10−8) for metabolite traits, stricter correlation thresholds (P<5×10−9) were implemented for selecting genetic IVs. These IVs were categorized based on the reference panel of Linkage Disequilibrium from the 1,000 Genomes Project (23,24), with a threshold of R2<0.001 at a distance of 1,000 kb. Due to the relatively small size of the GWAS dataset for metabolites, a cutoff (P=5×10−8) and a less stringent clustering threshold (R2<0.001 at a distance of 1,000 kb) were employed (19). To ensure the reliability of the tools used, IVs with F>10 were selected and identified as strong elements for subsequent analyses. Next, these IVs were obtained from the summary statistics pertaining to UGI cancer outcomes, excluding any that displayed potential pleiotropic effects (P<10−5) on UGI cancer, in line with methodologies from previous research (25). To maintain consistency in this analysis, discrepancies in SNPs between the exposure and outcome datasets were synchronized to ensure uniform effect estimates for the same effect allele (26).

Cell lines

The esophageal cancer cell line (KYSE150) and gastric cancer cell line (HGC27) selected for the experiment were purchased from the cell bank of the Chinese Academy of Sciences.

Main reagents and instruments

Cell culture medium and reagents included: Serum-free DMEM (cat. no. PM150210; Procell Life Science & Technology Co., Ltd.), RPMI 1640 basic medium (cat. no. PM150110; Procell Life Science & Technology Co., Ltd.), fetal bovine serum (FBS; cat. no. 164210-50; Procell Life Science & Technology Co., Ltd.), pancreatic enzyme (cat. no. PB180226; Procell Life Science & Technology Co., Ltd.), fructosyllysine (cat. no. HY-129380; MedChemExpress), 2′-deoxyuridine (cat. no. HY-D0186; MedChemExpress), cytidine (cat. no. HY-D0158; MedChemExpress), carnitine (cat. no. HY-B0399A; MedChemExpress), Cell Counting Kit-8 (CCK-8; Shanghai Biyuntian Biotechnology Co., Ltd.), ELISA reader (BioTek; Agilent Technologies, Inc.) and an inverted microscope (Nikon Corporation).

Cell culture

KYSE150 cells and HGC27 cells were incubated in DMEM and RPMI 1640 medium, respectively. The media were supplemented with 10% FBS and 100 U/ml penicillin and streptomycin (Gibco; Thermo Fisher Scientific, Inc.). The cells were maintained in an incubator set at 37°C with a 5% CO2 atmosphere. Upon reaching 80–90% confluence, the cells were enzymatically dissociated for subculturing.

Detection of cell proliferation ability

Cells were digested with 0.25% trypsin and resuspended for counting when entering the logarithmic growth phase. The cells were then seeded in a 96-well plate at a density of 3,000 cells/well. After attachment and morphological expansion, four common metabolites identified from MR results, namely fructosyllysine, 2′-deoxyuridine, cytidine and carnitine, were added for treatment at 37°C for 1.5 h in darkness. The concentrations of the metabolites were 0, 50 and 100 µM, with triplicate wells for each concentration. Subsequently, each well received 10 µl of CCK-8 reagent and was incubated at 37°C for 1.5 h in darkness. The optical density was measured at 450 nm using an ELISA reader at time intervals of 0, 24, and 48 h following the introduction of the metabolites.

Cell scratch assay

Cells were digested with 0.25% trypsin and counted during the logarithmic growth phase. The cells were then plated in a 6-well plate and each well contained 1.0×105 cells. Once the cells adhered and expanded morphologically, reaching ~80% confluence, a scratch was made on the cell monolayer using a 10-µl pipette tip. After making the scratch, the wells were rinsed twice with PBS to remove any detached cells and then a serum-free medium and varying concentrations of metabolites was added for continued incubation. Images of the scratch were captured at 0, 24 and 48 h after adding the metabolites using a microscope (Nikon Ts2FL inverted microscope; Nikon Corporation). The area of the scratch was quantified using ImageJ software (version 2023; National Institutes of Health). To assess the rate of cell migration and healing, the formula used for calculating wound healing rate was [(Original scratch area-Final scratch area)/Original scratch area] ×100%.

Statistical analysis

After extracting the data concerning SNPs associated with metabolites, including details such as effect alleles and their corresponding β values. The formula established previously (27) was used to calculate the genetic variance for each metabolite. All data were presented and the number of replicates performed following the reported formula (20). In the present study, a range of genetic variants were employed as IVs, rather than relying solely on an allele score. This approach was selected to thoroughly examine key assumptions, uncover potential pleiotropy and enhance the sensitivity efficacy of the multivariable MR analyses (28). A total of four distinct MR methodologies, including the inverse variance weighted (IVW; random-effects model), weighted median, MR-Egger and MR-PRESSO, were used to evaluate the consistency of the current findings under different assumptions about heterogeneity and pleiotropy. The IVW method, employing a random-effects model, served as the primary analysis framework for all four sets of IVs. Heterogeneity was quantified using Cochran's Q statistic.

The present study also included analyses with more stringent conditions. Assuming that all genetic variants are valid, the IVW method might be subject to bias if a considerable number of SNPs are influenced by horizontal pleiotropy (29). By contrast, the weighted median approach, effective when <50% of variants exhibit horizontal pleiotropy, operated under the assumption that the majority of genetic variants were valid (30). For situations where >50% of variants were affected by horizontal pleiotropy, the strength of the present genetic tools was evaluated through F statistics, considering a mean F<10 to be indicative of weak IVs (31).

Furthermore, the MR-Egger method was applied to assess potential directional pleiotropy, where a significant intercept would indicate a violation of IV assumptions, suggesting directional pleiotropy (32). In addition, the MR-PRESSO method was implemented to minimize heterogeneity in causal effect estimates by excluding disproportionately influential SNPs (NbDistribution=1,500) (33). In addition, Steiger-filtering was used to identify and exclude genetic variants more strongly linked to the outcome than to the exposure, indicative of potential reverse causality (34).

All statistical analyses in the present study were performed using R (R Foundation version 4.3.1; 2023 version R-project.org/) and specific R packages (‘TwoSampleMR’ and ‘MR’) (35) designed for MR analysis (36). The TwoSampleMR package facilitated the provision of causal estimates across the four MR models: IVW, weighted median, MR-Egger and MR-PRESSO. Analysis of functional experiment data was conducted using the software ImageJ (version 2023; National Institutes of Health) and GraphPad Prism 8 (GraphPad; Dotmatics). Unpaired Student's t-tests were used to calculate differences between the two groups. For group comparisons, one-way ANOVA and Bonferroni post hoc analysis were used. Data are expressed as the mean ± SD. P<0.05 was considered to indicate a statistically significant difference.

Results

Causal estimation between metabolites and UGI cancer
Causal estimation between metabolites and EC

The present study first assessed the causal impact of various metabolites on EC using the IVW method for a two-sample MR analysis. The assessment revealed that 57 metabolites were significantly associated with EC (Table I). Key findings highlighted notable associations with hub metabolites. Specifically, fructosyllysine levels showed a strong association with increased EC risk [odds ratio (OR)=1.450, 95% confidence interval (CI)=1.087–1.934, P=0.011]. Additionally, the ratio of 2′-deoxyuridine to cytidine (OR=1.464, 95% CI=1.111–1.929, P=0.007) and carnitine to protonylcarnitine (C3) (OR=0.655, 95% CI=0.499–0.861, P=0.002) were also significantly linked to EC risk (Fig. 2). Furthermore, a sensitivity analysis was conducted. Despite observing some heterogeneity and significant results from Cochran's Q test (P<0.05), the causal estimates remained robust under the random-effects IVW model. The P-values for the MR-Egger intercept were greater than 0.05, suggesting that no significant pleiotropy effects were found (Tables SI and SII). Additionally, the data were further assessed using scatter (Fig. 3A-C), funnel (Fig. 3D-F) and leave-one-out (Fig. 4A-C) plots and the potential influence of outliers and horizontal pleiotropy on the identified hub metabolites were excluded.

Table I.

Causal association between all metabolites and esophageal cancer. Inverse variance weighted was chosen as the primary method. P<0.05 was considered to indicate a statistically significant difference. OR >1 indicated a risk factor.

Table I.

Causal association between all metabolites and esophageal cancer. Inverse variance weighted was chosen as the primary method. P<0.05 was considered to indicate a statistically significant difference. OR >1 indicated a risk factor.

MetaboliteMethodNumber of single nucleotide polymorphismsP-valueOROR 95% lower CIOR 95% upper CI
Maltotriose levelsIVW240.0271.281.0271.603
Tartarate levelsIVW220.0290.730.5570.969
1-stearoyl-2-arachidonoyl-GPI (18:0/20:4) levelsIVW240.0041.341.0951.654
Stearidonate (18:4n3) levelsIVW270.0461.311.0041.726
Hexanoylglycine levelsIVW270.0361.201.0111.428
Beta-hydroxyisovaleroylcarnitine levelsIVW360.0270.760.6050.97
1-ribosyl-imidazoleacetate levelsIVW370.0100.750.6050.936
Thymol sulfate levelsIVW200.0481.341.0021.808
2-hydroxyhippurate levelsIVW200.0280.700.5200.964
2R,3R-dihydroxybutyrate levelsIVW320.0140.750.5990.946
1-(1-enyl-palmitoyl)-GPC (p-16:0) levelsIVW290.0211.321.0411.687
Docosadioate (C22-DC) levelsIVW260.0440.730.5470.992
N-palmitoylglycine levelsIVW220.0211.281.0361.582
Fructosyllysine levelsIVW270.0111.451.0871.934
Sphingomyelin (d18:2/14:0, d18: 1/14:1) levelsIVW240.0191.491.0672.098
Methyl-4-hydroxybenzoate sulfate levelsIVW220.0191.431.0601.936
1,2,3-benzenetriol sulfate (2) levelsIVW240.0030.650.4910.872
1,2-dilinoleoyl-GPC (18:2/18:2) levelsIVW170.0100.720.5630.927
1-palmitoyl-2-stearoyl-gpc (16:0/18:0) levelsIVW320.0181.291.0431.598
1-stearoyl-2-arachidonoyl-GPE (18:0/20:4) levelsIVW320.0061.241.0621.450
1-(1-enyl-palmitoyl)-2-arachidonoyl-GPE (p-16:0/20:4) levelsIVW220.0171.401.0601.850
1-palmitoyl-2-arachidonoyl-GPE (16:0/20:4) levelsIVW190.0441.181.0031.398
Docosahexaenoylcholine levelsIVW200.0490.720.5290.998
Sphingomyelin (d18:2/21:0, d16: 2/23:0) levelsIVW350.0221.341.0431.744
Sphingomyelin (d17:1/14:0, d16: 1/15:0) levelsIVW260.0101.431.0881.889
2,2′-Methylenebis(6-tert-butyl-p-cresol) levelsIVW210.0480.750.5660.998
3-hydroxy-2-methylpyridine sulfate levelsIVW210.0300.720.5480.969
4-acetylcatechol sulfate (1) levelsIVW190.0200.680.5020.943
Hydroxy-N6,N6,N6-trimethyllysine levelsIVW270.0291.351.0311.789
Alpha-tocopherol levelsIVW280.0161.401.0631.862
Beta-hydroxyisovalerate levelsIVW280.0331.311.0221.688
Eicosapentaenoate (EPA; 20:5n3) levelsIVW270.00051.541.2061.983
Cholesterol levelsIVW210.0181.441.0641.964
Gluconate levelsIVW240.0070.710.5530.911
Deoxycholate levelsIVW270.0321.321.0231.717
Ornithine levelsIVW280.0130.720.5630.936
Dimethylglycine levelsIVW330.0231.251.0311.525
Tyrosine levelsIVW290.0140.710.5390.934
N-stearoyl-sphinganine (d18:0/18:0) levelsIVW160.0030.640.4780.867
X-12101 levelsIVW250.0070.740.6040.926
X-22834 levelsIVW220.0410.730.5490.988
X-23639 levelsIVW230.0431.441.0112.057
X-23648 levelsIVW190.0241.411.0451.927
X-23641 levelsIVW300.0010.710.5800.872
X-23665 levelsIVW200.0480.700.4950.997
X-23739 levelsIVW280.0110.750.6040.938
X-25810 levelsIVW360.0290.770.6190.974
3-methylcytidine levelsIVW200.0280.830.7170.981
Adenosine 5′-diphosphate to Adenosine 5′-monophosphate ratioIVW260.0181.271.0421.557
Cortisone to cortisol ratioIVW200.0080.640.460.892
Phosphate to N-palmitoyl-sphingosine (d18:1 to 16:0) ratioIVW240.0490.750.5711.000
Adenosine 5′-monophosphate to arginine ratioIVW170.0320.650.440.964
2′-deoxyuridine to cytidine ratioIVW300.0071.461.0891.751
Acetylcarnitine to propionylcarnitine ratioIVW190.0380.710.5220.982
Carnitine to propionylcarnitine ratioIVW240.0020.660.4990.861
Alpha-ketoglutarate to trans-4-hydroxyproline ratioIVW200.0201.421.0551.936
Adenosine 5′-monophosphate to urate ratioIVW270.0201.411.0541.896

[i] OR, odds ratio; IVW, inverse variance weighted; CI, confidence interval.

Causal estimation between metabolites and GC

Using the IVW method as the primary analytic approach, a two-sample MR analysis was adopted to investigate the causal effect of GC on metabolites. The results identified associations with 58 metabolites (Table II). Notably, the GC risk was observed to be significantly associated with elevated levels of fructosyllysine (OR=1.70, 95% CI=1.240–2.346, P=0.001), the 2′-deoxyuridine to cytidine ratio (OR=1.464, 95% CI=1.094–1.957, P=0.010) and the C3 ratio (OR=0.664, 95% CI=0.486–0.906, P=0.010; Fig. 5). Although sensitivity analysis highlighted some heterogeneity, Cochran's Q yielded P<0.05 and the causality estimates were satisfactory when using a random-effects IVW approach. The P-values for the MR-Egger intercept were >0.05, indicating that no significant pleiotropy effects were found (Tables SIII and SIV). Additionally, scatter (Fig. 6A-C), funnel (Fig. 6D-F) and leave-one-out (Fig. 7A-C) plots excluded the likelihood inflicting by potential outliers and horizontal pleiotropy on the identified hub metabolites that were excluded.

Table II.

The causal association between all metabolites and gastric cancer. Inverse variance weighted was chosen as the primary method. P<0.05 was considered to indicate a statistically significant difference. OR >1 indicated a risk factor.

Table II.

The causal association between all metabolites and gastric cancer. Inverse variance weighted was chosen as the primary method. P<0.05 was considered to indicate a statistically significant difference. OR >1 indicated a risk factor.

MetaboliteMethodNumber of single nucleotide polymorphismsP-valueOROR 95% lower CIOR 95% upper CI
Gentisate levelsIVW270.0361.441.0242.028
1,5-anhydroglucitol (1,5-ag) levelsIVW320.0181.351.0521.742
2-linoleoylglycerol (18:2) levelsIVW230.0161.431.0691.921
3-carboxy-4-methyl-5-propyl-2-furanpropanoate (cmpf) levelsIVW170.0471.481.0042.201
10-nonadecenoate (19:1n9) levelsIVW130.0180.570.3630.912
Stachydrine levelsIVW240.0361.411.0211.961
2-palmitoleoyl-GPC (16:1) levelsIVW260.0451.331.0061.769
Isobutyrylglycine levelsIVW300.0271.31.0291.666
Glycerophosphoethanolamine levelsIVW250.0481.341.0021.807
Taurolithocholate 3-sulfate levelsIVW230.0180.690.5100.939
Glycolithocholate sulfate levelsIVW230.00090.590.4350.808
Pregnenolone sulfate levelsIVW360.0230.720.5430.955
S-methylmethionine levelsIVW210.0180.680.5000.939
2,3-dihydroxyisovalerate levelsIVW270.0021.611.1852.212
N-oleoyltaurine levelsIVW210.0081.461.1031.956
2-aminooctanoate levelsIVW290.0371.231.0111.495
3-acetylphenol sulfate levelsIVW210.0371.431.0202.018
N-acetylalliin levelsIVW300.0010.690.5570.872
Fructosyllysine levelsIVW270.0011.701.2002.483
N-acetyltaurine levelsIVW200.0450.710.5160.992
3-hydroxypyridine sulfate levelsIVW240.0141.501.0842.081
Arabitol/xylitol levelsIVW240.0490.700.4930.999
Tricosanoyl sphingomyelin (d18: 1/23:0) levelsIVW290.0060.640.4690.882
1-stearoyl-2-arachidonoyl-GPE (18:0/20:4) levelsIVW320.0121.331.0621.668
5-hydroxyindole sulfate levelsIVW190.0491.361.0011.864
Nisinate (24:6n3) levelsIVW180.0031.511.1452.002
Arachidonoylcarnitine (C20:4) levelsIVW350.0041.271.0761.508
Heptenedioate (C7:1-DC) levelsIVW160.0161.691.1032.605
Octadecenedioate (C18:1-DC) levelsIVW230.0130.750.6060.943
Perfluorooctanoate (PFOA) levelsIVW230.0360.690.4910.977
Glyco-beta-muricholate levelsIVW250.0170.760.6170.954
8-methoxykynurenate levelsIVW260.0261.381.0381.850
2-ketocaprylate levelsIVW230.0281.371.0341.837
Palmitoyl-sphingosine-phosphoe-thanolamine (d18:1/16:0) levelsIVW290.0050.660.4960.887
Picolinate levelsIVW230.0320.740.5650.975
Erucate (22:1n9) levelsIVW190.0030.580.4100.842
Cys-gly, oxidized levelsIVW260.0281.331.0301.719
1-palmitoyl-2-linoleoyl-gpc (16:0/18:2) levelsIVW350.0400.780.6220.989
Eicosapentaenoate (20:5n3) levelsIVW270.0401.341.0121.791
Stearate (18:0) levelsIVW210.0330.670.4690.97
Pseudouridine levelsIVW230.0040.590.4180.85
X-11632 levelsIVW260.0140.680.5080.928
X-12701 levelsIVW150.0060.60.4260.871
X-18901 levelsIVW300.0061.51.1192.018
X-21283 levelsIVW170.0070.720.580.917
X-22834 levelsIVW220.0260.710.5250.961
X-23648 levelsIVW190.0281.481.0432.113
X-23782 levelsIVW200.0271.571.0512.365
X-24344 levelsIVW170.0061.541.1292.116
X-24585 levelsIVW190.0131.581.0992.29
Oleoyl-linoleoyl-glycerol (18:1 to 18:2) to linoleoyl-arachidonoyl-glycerol (18:2 to 20:4) ratioIVW250.0210.800.6610.968
Spermidine to adenosine 5′-diphosphate (ADP) ratioIVW250.0390.780.6240.987
Glycine to phosphate ratioIVW280.0241.271.0321.565
Adenosine 5′-diphosphate (ADP) to glutamate ratioIVW320.0220.770.6190.963
Cortisol to 4-cholesten-3-one ratioIVW210.0201.531.0682.192
2′-deoxyuridine to cytidine ratioIVW300.0101.461.0941.957
Carnitine to propionylcarnitine (C3) ratioIVW240.0100.660.4860.906
Arachidonate (20:4n6) to linoleate (18:2n6) ratioIVW260.0171.401.0621.865

[i] OR, odds ratio; IVW, inverse variance weighted.

Hub metabolites affect the proliferation and migration abilities of EC and GC cells

To verify the effects of four metabolites, fructosyllysine, 2′-deoxyuridine, cytidine and carnitine, on the proliferation and migration abilities of EC and GC cells, the present study established in vitro models of EC and GC using KYSE150 and HGC27 cells, respectively. CCK-8 assay showed that fructosyllysine and 2′-deoxyuridine promoted the proliferation of EC and GC cells, while cytidine and carnitine exhibited inhibitory effects on the proliferation of these two tumor cell lines. Thus, the 2′-deoxyuridine to cytidine ratio was associated with the proliferation promotion of EC and GC cells. Scratch assay demonstrated that EC and GC cells treated with fructosyllysine and 2′-deoxyuridine showed significantly faster scratch healing compared with the control group, indicating enhanced migratory capabilities. By contrast, cells treated with cytidine or carnitine displayed notably slower wound healing than those of the control group, suggesting a suppressive effect on cell migration. Thus, the scratch healing speed of EC and GC cells treated with a ratio of 2′-deoxyuridine to cytidine was significantly faster compared with the control group. In summary, the metabolites fructosyllysine and the 2′-deoxyuridine to cytidine ratio were found to enhance the proliferation and migration of EC and GC cells. Conversely, carnitine was observed to inhibit both the proliferation and migration of these tumor cell types (Fig. 8, Fig. 9, Fig. 10, Fig. 11).

Discussion

MR analysis is increasingly recognized for its ability to delineate potential causal relationships between risk factors and diseases (19). The present study identified 57 metabolites associated with EC and 58 with GC. Notably, fructosyllysine levels, 2′-deoxyuridine to cytidine ratio and C3 ratio emerged as common metabolites influencing EC and GC. Among them, fructosyllysine levels and 2′-deoxyridine to cytidine ratio were identified as risk factors for both cancers, while the C3 ratio appeared as protective factors for GC and EC.

Fructosyllysine is a precursor of carboxymethyllysine, an advanced glycation end-product (AGE) presenting in heated foods, which is considered potentially harmful to human health (37). AGEs are produced through non-enzymatic glycation reactions between amino acids and reducing sugars, a process commonly known as the Maillard reaction (38). AGEs exist in both protein-bound and free forms in food, with the majority being absorbed through the gastrointestinal tract. According to previous research, AGEs contribute to the progression of various chronic diseases, including diabetes-related complications, cardiovascular, renal and neurodegenerative disorders, as well as aging (3841). Carboxymethyllysine, the predominant component of AGEs, is formed through fructosyllysine oxidation (42), which is linked to inflammation and endothelial dysfunction. Notably, chronic inflammation in the gastrointestinal tract is closely associated with cancer (43). AGEs primarily mediate inflammation by binding to the receptor for AGE (RAGE). Expression of RAGE has been detected in human intestinal epithelial cells and colon tissues, where its engagement with ligands enhances the oxidase activity of nicotinamide adenine dinucleotide phosphate, leading to the transcription of NF-κB and the subsequent expression of pro-inflammatory genes, such as IL-6 and TNF-α (44). Thus, an elevated level of fructosyllysine may trigger the inflammatory response in the intestine, potentially inducing cancer over time. In the study by Raupbach et al (45), the expression of RAGE in colon cancer HCT116 cells was shown to be comparable to that in the intestinal epithelium and the absorption efficacy of fructosyllysine by HCT116 cells was largely related to their structural characteristics. However, reports linking fructosyllysine to cancer are limited. In the present study, increased fructosyllysine levels were identified as a risk factor for EC and GC, consistent with previous findings (44,45), although the mechanisms require further exploration, particularly concerning the role of inflammatory activation. Additionally, previous studies have shown that gut microbiota can degrade AGEs and their precursors under anaerobic conditions, reducing the exposure to dietary fructosyllysine or carboxymethyllysine in the gastrointestinal tract and thereby mitigating potential harm to humans (37). Investigating the role of gut microbiota presents a promising avenue for developing treatments for gastrointestinal tumors.

The present study indicated an increase in the 2′-deoxyuridine to cytidine ratio as a risk factor for GC and EC. Cytidine deaminase (CDA) is an enzyme involved in the pyrimidine recovery pathway, catalyzing the hydrolysis and deamination of cytidine and 2′-deoxycytidine to produce uridine and 2′-deoxyuridine (46). Research has demonstrated that CDA expression is often lost or downregulated in various types of cancer in breast, lung, ovarian, liver, colon and cervix, including EC (47). Low levels of CDA may contribute to cancer development (48) by suppressing the deoxyuridine synthesis. Deoxyuridine plays a role in mitigating reactive oxygen species (ROS)-induced endoplasmic reticulum stress, thereby supporting cancer cell survival; thus, its increased synthesis could potentially facilitate tumor development (49). In GC and EC, antitumor therapy commonly results in the accumulation of ROS, which promotes cancer cell apoptosis and inhibits tumor growth (5053). The present results suggested that the 2′-deoxyuridine to cytidine ratio was a risk factor for EC and GC.

Carnitine serves as a potent antioxidant with multiple regulatory functions, such as boosting cellular respiration, promoting apoptosis and decreasing both tumor cell growth and inflammation. L-carnitine, the active form of carnitine, transports long-chain fatty acid acyl groups from the cytoplasm into the mitochondrial matrix. This transport facilitates the β-oxidation process, converting these acids into acetyl-coenzyme A, which then enters the citric acid cycle to produce energy. However, tumor cells proliferate rapidly and require notable energy and they rely on a distinct metabolic pathway known as the Warburg effect (54), but not the citric acid cycle, to proliferate. The accumulation of carnitine can promote the citric acid cycle, creating an environment that is harmful to the proliferation of cancer cells and even affects their stemness (55). Acylcarnitine is an ester produced by the combination of fatty acid (that is. acyl) and L-carnitine, which plays an important role in numerous cell metabolism pathways related to energy production. Acylcarnitine has been identified as an important indicator of multiple metabolism-related diseases, including cardiovascular diseases, diabetes, metabolic disorders, depression and certain tumors (56). Short-chain acylcarnitine is the most abundant acylcarnitine class in the human body and it has been reported to be closely associated with mental illness (57). Propionyl-L-carnitine is a type of carnitine donor that increases the transport of fatty acids across mitochondrial membranes and is used as a supplement for the treatment of depression (56). In the study by Zhang et al (58), a negative correlation between propionyl-L-carnitine and lung cancer was observed. In addition, L-carnitine has also been shown to improve cancer-related anorexia/cachexia by reducing inflammation (59). Agreeing with previous research, an increase in the ratio of C3 may promote the citric acid cycle, inhibit tumor cell proliferation and serve as a co-protective factor for EC and GC. The functional experiments in the present study also demonstrated that carnitine inhibited tumor cell proliferation.

The current two-sample MR analysis was designed to evaluate the causal relationship between metabolites and UGI tumors using large-sample GWAS and UK Biobank data. This approach overcame some limitations of traditional observational studies by reducing the influence of confounding factors and reverse causality. In addition, MR mitigated the representativeness and feasibility issues that are common in RCTs. However, the present study faced several challenges. First, non-fasting plasma samples were used for metabolomics profiling. Despite adjustments made for the time elapsed since the last meal or beverage, residual variability may still influence the results. Second, the study focused on gene-metabolite pairs identified from current expression data and biological understanding, specifically targeting effector genes. Nonetheless, the potential relevance of other highly heritable metabolites or ratios to the disease cannot be overlooked. Future studies should expand on this by including additional expression data and metabolic insights to identify effector genes for these additional metabolites and ratios. Third, the MR analyses used in the present study were constrained by the fact that most metabolites and metabolite ratios were associated with only a single IV. This constraint made it difficult to apply common MR sensitivity tests such as MR-Egger, which require multiple IVs. However, the selection of IVs that are closely linked to effector genes influencing metabolite levels in the present study mitigated the risk of horizontal pleiotropy. Additionally, assessments of metabolic pleiotropy were conducted to remove IVs linked to multiple metabolites not involved in the same metabolic pathways. Despite these measures, biases may not be fully eliminated due to limitations in metabolome profiling and uncertainties in metabolite-protein interaction databases. Finally, the present study primarily involved elderly individuals of European descent. Future research should explore the effects of identified genetic variations on metabolites and their ratios in a more diverse demographic spectrum. Moreover, the current study covered a broad range of metabolites; however, the metabolic pathway and mechanisms of certain metabolites in disease remain unclear, which limited the interpretability of MR findings.

In summary, this comprehensive MR analysis of metabolites has uncovered new gene-metabolite associations, offering an enhanced understanding of genetic regulation in human metabolism. These discoveries are expected to facilitate the identification of potential markers and targets that could aid in precision prevention and lead to behavioral modifications, innovative pharmaceutical interventions and personalized treatments. Such strategies would be specifically designed to address the unique metabolic vulnerabilities associated with UGI tumors.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by the Natural Science Foundation of Guangdong Province (grant no. 2023A1515012548) and the Science and Technology Program of Guangzhou (grant no. 2024A04J4802).

Availability of data and materials

The data generated in the present study may be found in the GWAS catalog and UKBiobank ICD PheWeb under accession number (GCST90199621-GCST90201020, inclusive; ukb-saige-150; ukb-saige-151) or at the following URL: (https://www.ebi.ac.uk/gwas/ and https://pheweb.org/UKB-SAIGE/, or https://pheweb.org/UKB-SAIGE/pheno/150 and http://pheweb.org/UKB-SAIGE/pheno/151).

Authors' contributions

PNo collected, analyzed and interpreted the data. contributed to conception, design and drafted the manuscript. CZ performed the cell scratch assay and the CCK-8 assay. SS, ST, SK, PNi, QK, YL, AP, WF, KD and JL designed, revised and supervised the study. PNo and KD confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024. View Article : Google Scholar : PubMed/NCBI

2 

Zong L, Abe M, Seto Y and Ji J: The challenge of screening for early gastric cancer in China. Lancet. 388:26062016. View Article : Google Scholar : PubMed/NCBI

3 

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

4 

Huang S, Guo Y, Li Z, Zhang Y, Zhou T, You W, Pan K and Li W: A systematic review of metabolomic profiling of gastric cancer and esophageal cancer. Cancer Biol Med. 17:181–198. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Abbassi-Ghadi N, Kumar S, Huang J, Goldin R, Takats Z and Hanna GB: Metabolomic profiling of oesophago-gastric cancer: A systematic review. Eur J Cancer. 49:3625–3637. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Gu J, Hu X, Shao W, Ji T, Yang W, Zhuo H, Jin Z, Huang H, Chen J, Huang C and Lin D: Metabolomic analysis reveals altered metabolic pathways in a rat model of gastric carcinogenesis. Oncotarget. 7:60053–60073. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Kim KB, Yang JY, Kwack SJ, Park KL, Kim HS, Ryu DH, Kim YJ, Hwang GS and Lee BM: Toxicometabolomics of urinary biomarkers for human gastric cancer in a mouse model. J Toxicol Environ Health A. 73:1420–1430. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Matsunaga S, Nishiumi S, Tagawa R and Yoshida M: Alterations in metabolic pathways in gastric epithelial cells infected with Helicobacter pylori. Microb Pathog. 124:122–129. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Di Gialleonardo V, Tee SS, Aldeborgh HN, Miloushev VZ, Cunha LS, Sukenick GD and Keshari KR: High-throughput indirect quantitation of 13C enriched metabolites using 1H NMR. Anal Chem. 88:11147–11153. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Holmes E, Wilson ID and Nicholson JK: Metabolic phenotyping in health and disease. Cell. 134:714–717. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Wishart DS: Emerging applications of metabolomics in drug discovery and precision medicine. Nat Rev Drug Discov. 15:473–484. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Chan AW, Gill RS, Schiller D and Sawyer MB: Potential role of metabolomics in diagnosis and surveillance of gastric cancer. World J Gastroenterol. 20:12874–12882. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Che J, Zhao Y, Gu B, Li S, Li Y, Pan K, Sun T, Han X, Lv J, Zhang S, et al: Untargeted serum metabolomics reveals potential biomarkers and metabolic pathways associated with the progression of gastroesophageal cancer. BMC Cancer. 23:12382023. View Article : Google Scholar : PubMed/NCBI

14 

Tokunaga M, Kami K, Ozawa S, Oguma J, Kazuno A, Miyachi H, Ohashi Y, Kusuhara M and Terashima M: Metabolome analysis of esophageal cancer tissues using capillary electrophoresis-time-of-flight mass spectrometry. Int J Oncol. 52:1947–1958. 2018.PubMed/NCBI

15 

Wang J, Kunzke T, Prade VM, Shen J, Buck A, Feuchtinger A, Haffner I, Luber B, Liu DHW, Langer R, et al: A serum metabolomics analysis reveals a panel of screening metabolic biomarkers for esophageal squamous cell carcinoma. Clin Transl Med. 11:e4192021. View Article : Google Scholar : PubMed/NCBI

16 

Li J, Li S, Yu L, Wei J, Sun H, Yang C and Tan H: Identification of preoperative serum metabolites associated with postoperative opioid consumption in gastric cancer patients by extreme phenotype sampling. Pain Physician. 25:E385–E396. 2022.PubMed/NCBI

17 

Yuan LW, Yamashita H and Seto Y: Glucose metabolism in gastric cancer: The cutting-edge. World J Gastroenterol. 22:2046–2059. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Xiao S and Zhou L: Gastric cancer: Metabolic and metabolomics perspectives (review). Int J Oncol. 51:5–17. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Davey Smith G and Hemani G: Mendelian randomization: Genetic anchors for causal inference in epidemiological studies. Hum Mol Genet. 23((R1)): R89–R98. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Chen Y, Lu T, Pettersson-Kymmer U, Stewart ID, Butler-Laporte G, Nakanishi T, Cerani A, Liang KYH, Yoshiji S, Willett JDS, et al: Genomic atlas of the plasma metabolome prioritizes metabolites implicated in human diseases. Nat Genet. 55:44–53. 2023. View Article : Google Scholar : PubMed/NCBI

21 

Burgess S and Thompson SG: Interpreting findings from Mendelian randomization using the MR-egger method. Eur J Epidemiol. 32:377–389. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Feng Q, Lacey B, Bešević J, Omiyale W, Conroy M, Starkey F, Calvin C, Callen H, Bramley L, Welsh S, et al: UK biobank: Enhanced assessment of the epidemiology and long-term impact of coronavirus disease-2019. Camb Prism Precis Med. 1:e302023. View Article : Google Scholar : PubMed/NCBI

23 

Yang J, Yan B, Zhao B, Fan Y, He X, Yang L, Ma Q, Zheng J, Wang W, Bai L, et al: Assessing the causal effects of human serum metabolites on 5 major psychiatric disorders. Schizophr Bull. 46:804–813. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Choi KW, Chen CY, Stein MB, Klimentidis YC, Wang MJ, Koenen KC and Smoller JW; Major Depressive Disorder Working Group of the Psychiatric Genomics Consortium, : Assessment of bidirectional relationships between physical activity and depression among adults: A 2-sample mendelian randomization study. JAMA Psychiatry. 76:399–408. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Sun Y, Zhou J and Ye K: White blood cells and severe COVID-19: A mendelian randomization study. J Pers Med. 11:1952021. View Article : Google Scholar : PubMed/NCBI

26 

Sidore C, Busonero F, Maschio A, Porcu E, Naitza S, Zoledziewska M, Mulas A, Pistis G, Steri M, Danjou F, et al: Genome sequencing elucidates Sardinian genetic architecture and augments association analyses for lipid and blood inflammatory markers. Nat Genet. 47:1272–1281. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Park JH, Wacholder S, Gail MH, Peters U, Jacobs KB, Chanock SJ and Chatterjee N: Estimation of effect size distribution from genome-wide association studies and implications for future discoveries. Nat Genet. 42:570–575. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Davies NM, Holmes MV and Davey SG: Reading Mendelian randomisation studies: A guide, glossary, and checklist for clinicians. BMJ. 362:k6012018. View Article : Google Scholar : PubMed/NCBI

29 

Hartwig FP, Davey Smith G and Bowden J: Robust inference in summary data Mendelian randomization via the zero modal pleiotropy assumption. Int J Epidemiol. 46:1985–1998. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Bowden J, Davey Smith G, Haycock PC and Burgess S: Consistent estimation in Mendelian randomization with some invalid instruments using a weighted median estimator. Genet Epidemiol. 40:304–314. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Allen RJ, Porte J, Braybrooke R, Flores C, Fingerlin TE, Oldham JM, Guillen-Guio B, Ma SF, Okamoto T, John AE, et al: Genetic variants associated with susceptibility to idiopathic pulmonary fibrosis in people of European ancestry: A genome-wide association study. Lancet Respir Med. 5:869–880. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Burgess S, Bowden J, Fall T, Ingelsson E and Thompson SG: Sensitivity analyses for robust causal inference from Mendelian randomization analyses with multiple genetic variants. Epidemiology. 28:30–42. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Verbanck M, Chen CY, Neale B and Do R: Detection of widespread horizontal pleiotropy in causal relationships inferred from Mendelian randomization between complex traits and diseases. Nat Genet. 50:693–698. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Hemani G, Tilling K and Davey Smith G: Orienting the causal relationship between imprecisely measured traits using GWAS summary data. PLoS Genet. 13:e10070812017. View Article : Google Scholar : PubMed/NCBI

35 

Yavorska OO and Burgess S: MendelianRandomization: An R package for performing Mendelian randomization analyses using summarized data. Int J Epidemiol. 46:1734–1739. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Hemani G, Zheng J, Elsworth B, Wade KH, Haberland V, Baird D, Laurin C, Burgess S, Bowden J, Langdon R, et al: The MR-base platform supports systematic causal inference across the human phenome. Elife. 7:e344082018. View Article : Google Scholar : PubMed/NCBI

37 

van Dongen KCW, Belzer C, Bakker W, Rietjens IMCM and Beekmann K: Inter- and intraindividual differences in the capacity of the human intestinal microbiome in fecal slurries to metabolize fructoselysine and carboxymethyllysine. J Agric Food Chem. 70:11759–11768. 2022. View Article : Google Scholar : PubMed/NCBI

38 

Sergi D, Boulestin H, Campbell FM and Williams LM: The role of dietary advanced glycation end products in metabolic dysfunction. Mol Nutr Food Res. 65:e19009342021. View Article : Google Scholar : PubMed/NCBI

39 

Rabbani N and Thornalley PJ: Hidden complexities in the measurement of fructosyl-lysine and advanced glycation end products for risk prediction of vascular complications of diabetes. Diabetes. 64:9–11. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Sobenin IA, Tertov VV, Koschinsky T, Bünting CE, Slavina ES, Dedov II and Orekhov AN: Modified low density lipoprotein from diabetic patients causes cholesterol accumulation in human intimal aortic cells. Atherosclerosis. 100:41–54. 1993. View Article : Google Scholar : PubMed/NCBI

41 

Ahmed N, Ahmed U, Thornalley PJ, Hager K, Fleischer G and Münch G: Protein glycation, oxidation and nitration adduct residues and free adducts of cerebrospinal fluid in Alzheimer's disease and link to cognitive impairment. J Neurochem. 92:255–263. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Ahmed MU, Thorpe SR and Baynes JW: Identification of N epsilon-carboxymethyllysine as a degradation product of fructoselysine in glycated protein. J Biol Chem. 261:4889–4894. 1986. View Article : Google Scholar : PubMed/NCBI

43 

Li L, Liu H, Yu J, Sun Z, Jiang M, Yu H and Wang C: Intestinal microbiota and metabolomics reveal the role of auricularia delicate in regulating colitis-associated colorectal cancer. Nutrients. 15:50112023. View Article : Google Scholar : PubMed/NCBI

44 

Zen K, Chen CXJ, Chen YT, Wilton R and Liu Y: Receptor for advanced glycation endproducts mediates neutrophil migration across intestinal epithelium. J Immunol. 178:2483–2490. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Raupbach J, Müller SK, Schnell V, Friedrich S, Hellwig A, Grune T and Henle T: The effect of free and protein-bound maillard reaction products N-ε-carboxymethyllysine, N-ε-fructosyllysine, and pyrraline on Nrf2 and NFκB in HCT 116 cells. Mol Nutr Food Res. 67:e23001372023. View Article : Google Scholar : PubMed/NCBI

46 

Urbelienė N, Tiškus M, Tamulaitienė G, Gasparavičiūtė R, Lapinskaitė R, Jauniškis V, Sūdžius J, Meškienė R, Tauraitė D, Skrodenytė E, et al: Cytidine deaminases catalyze the conversion of N(S,O)4-substituted pyrimidine nucleosides. Sci Adv. 9:eade43612023. View Article : Google Scholar : PubMed/NCBI

47 

Chen Y, Ma Z, Shen X, Li L, Zhong J, Min LS, Xu L, Li H, Zhang J and Dai L: Serum lipidomics profiling to identify biomarkers for non-small cell lung cancer. Biomed Res Int. 2018:52762402018.PubMed/NCBI

48 

Onclercq-Delic R, Buhagiar-Labarchède G, Leboucher S, Larcher T, Ledevin M, Machon C, Guitton J and Amor-Guéret M: Cytidine deaminase deficiency in mice enhances genetic instability but limits the number of chemically induced colon tumors. Cancer Lett. 555:2160302023. View Article : Google Scholar : PubMed/NCBI

49 

Olou AA, King RJ, Yu F and Singh PK: MUC1 oncoprotein mitigates ER stress via CDA-mediated reprogramming of pyrimidine metabolism. Oncogene. 39:3381–3395. 2020. View Article : Google Scholar : PubMed/NCBI

50 

Deng YQ, Gao M, Lu D, Liu QP, Zhang RJ, Ye J, Zhao J, Feng ZH, Li QZ and Zhang H: Compound-composed Chinese medicine of Huachansu triggers apoptosis of gastric cancer cells through increase of reactive oxygen species levels and suppression of proteasome activities. Phytomedicine. 123:1551692024. View Article : Google Scholar : PubMed/NCBI

51 

Sun W, Yuan Y, Chen J, Bao Q, Shang M, Sun P and Peng H: Construction and validation of a novel senescence-related risk score can help predict the prognosis and tumor microenvironment of gastric cancer patients and determine that STK40 can affect the ROS accumulation and proliferation ability of gastric cancer cells. Front Immunol. 14:12592312023. View Article : Google Scholar : PubMed/NCBI

52 

Javid H, Hashemy SI, Heidari MF, Esparham A and Gorgani-Firuzjaee S: The anticancer role of cerium oxide nanoparticles by inducing antioxidant activity in esophageal cancer and cancer stem-like ESCC spheres. Biomed Res Int. 2022:32681972022. View Article : Google Scholar : PubMed/NCBI

53 

Cannon A, Maher SG and Lynam-Lennon N: Generation and characterization of an isogenic cell line model of radioresistant esophageal adenocarcinoma. Methods Mol Biol. 2645:139–152. 2023. View Article : Google Scholar : PubMed/NCBI

54 

DeBerardinis RJ: Is cancer a disease of abnormal cellular metabolism? New angles on an old idea. Genet Med. 10:767–777. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Farahzadi R, Sanaat Z, Movassaghpour-Akbari AA, Fathi E and Montazersaheb S: Investigation of L-carnitine effects on CD44+ cancer stem cells from MDA-MB-231 breast cancer cell line as anti-cancer therapy. Regen Ther. 24:219–226. 2023. View Article : Google Scholar : PubMed/NCBI

56 

Dambrova M, Makrecka-Kuka M, Kuka J, Vilskersts R, Nordberg D, Attwood MM, Smesny S, Sen ZD, Guo AC, Oler E, et al: Acylcarnitines: Nomenclature, biomarkers, therapeutic potential, drug targets, and clinical trials. Pharmacol Rev. 74:506–551. 2022. View Article : Google Scholar : PubMed/NCBI

57 

Nasca C, Bigio B, Lee FS, Young SP, Kautz MM, Albright A, Beasley J, Millington DS, Mathé AA, Kocsis JH, et al: Acetyl-l-carnitine deficiency in patients with major depressive disorder. Proc Natl Acad Sci USA. 115:8627–8632. 2018. View Article : Google Scholar : PubMed/NCBI

58 

Zhang X, Wang C, Li C and Zhao H: Development and internal validation of nomograms based on plasma metabolites to predict non-small cell lung cancer risk in smoking and nonsmoking populations. Thorac Cancer. 14:1719–1731. 2023. View Article : Google Scholar : PubMed/NCBI

59 

Laviano A, Molfino A, Seelaender M, Frascaria T, Bertini G, Ramaccini C, Bollea MR, Citro G and Rossi Fanelli F: Carnitine administration reduces cytokine levels, improves food intake, and ameliorates body composition in tumor-bearing rats. Cancer Invest. 29:696–700. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2024
Volume 30 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Nov P, Zheng C, Wang D, Sou S, Touch S, Kouy S, Ni P, Kou Q, Li Y, Prasai A, Prasai A, et al: Causal association between metabolites and upper gastrointestinal tumors: A Mendelian randomization study. Mol Med Rep 30: 212, 2024.
APA
Nov, P., Zheng, C., Wang, D., Sou, S., Touch, S., Kouy, S. ... Li, J. (2024). Causal association between metabolites and upper gastrointestinal tumors: A Mendelian randomization study. Molecular Medicine Reports, 30, 212. https://doi.org/10.3892/mmr.2024.13336
MLA
Nov, P., Zheng, C., Wang, D., Sou, S., Touch, S., Kouy, S., Ni, P., Kou, Q., Li, Y., Prasai, A., Fu, W., Du, K., Li, J."Causal association between metabolites and upper gastrointestinal tumors: A Mendelian randomization study". Molecular Medicine Reports 30.6 (2024): 212.
Chicago
Nov, P., Zheng, C., Wang, D., Sou, S., Touch, S., Kouy, S., Ni, P., Kou, Q., Li, Y., Prasai, A., Fu, W., Du, K., Li, J."Causal association between metabolites and upper gastrointestinal tumors: A Mendelian randomization study". Molecular Medicine Reports 30, no. 6 (2024): 212. https://doi.org/10.3892/mmr.2024.13336