Abstract
Trigonelline is a phytoalkaloid commonly found in green and roasted coffee beans. It is also found in decaffeinated coffee. Previous report has shown that extract from trigonelline-rich plant exhibits anti-lithiatic effects in a nephrolithiatic rat model. Nevertheless, cellular mechanisms underlying the anti-lithiatic properties of trigonelline remain hazy. Herein, we used nanoLC-ESI-Qq-TOF MS/MS and MaxQuant-based quantitative proteomics to identify trigonelline-induced changes in protein expression in MDCK renal cells. From a total of 1006 and 1011 proteins identified from control and trigonelline-treated cells, respectively, levels of 62 (23 upregulated and 39 downregulated) proteins were significantly changed by trigonelline. Functional enrichment and reactome pathway analyses suggested that these 62 altered proteins were related to stress response, cell cycle and cell polarity. Functional validation by corresponding experimental assays revealed that trigonelline prevented calcium oxalate monohydrate crystal-induced renal cell deteriorations by inhibiting crystal-induced overproduction of intracellular reactive oxygen species, G0/G1 to G2/M cell cycle shift, tight junction disruption, and epithelial-mesenchymal transition. These findings provide cellular mechanisms and convincing evidence for the renoprotective effects of trigonelline, particularly in kidney stone prevention.
Keywords: Anti-lithiatic, Bioactive compound, Bioinformatics, Coffee, Kidney stone, Proteomics, Reactome
Graphical Abstract

Highlights
-
•
Trigonelline induces altered levels of 62 proteins in MDCK renal cells.
-
•
The altered proteins are related to stress response, cell cycle and cell polarity.
-
•
COM crystals induce oxidative stress and cell cycle shift from G0/G1 to G2/M.
-
•
COM crystals disrupt tight junction and cause epithelial-mesenchymal transition.
-
•
All of these COM-induced defects can be prevented by trigonelline pretreatment.
1. Introduction
Trigonelline is a bioactive phytoalkaloid predominantly found in a traditional herb namely fenugreek or Trigonella foenum-graecum [1], [2]. This compound is also found in green and roasted coffee beans [3]. Arabica green coffee contains trigonelline at 9.87 – 12.60 mg/g [4], whereas Arabica expresso and Robusta coffee contain trigonelline at 70 mg/cup and 40 mg/cup, respectively [5]. Roasting condition also affects trigonelline content, e.g., fast-roasting process better preserves trigonelline content than slow-roasting procedure [6], [7]. Roasting at temperature < 225 ℃ is recommended to enrich trigonelline content because the higher roasting temperature causes trigonelline degradation [3]. Trigonelline content in the coffee roasted at 225 ℃ for 120 s is approximately 0.80 g/100 g dry weight [3]. Interestingly, trigonelline is not much affected by decaffeinated process, as it is slightly reduced from 0.80 to 0.69 g/100 g dry weight after decaffeination [3].
Serum and urinary trigonelline levels can serve as biomarkers for coffee consumption [8], [9], [10]. The potential benefits of trigonelline have been revealed in several diseases, such as Alzheimer’s disease [11], [12], [13] and diabetes [11]. Kidney stone disease (also known as nephrolithiasis/urolithiasis) is a result of deposition of crystalline compounds packed as the solid mass within the kidney. Among the crystalline compounds, calcium oxalate monohydrate (COM) is the predominant type found in clinical stones [14], [15]. The in vivo anti-lithiatic effects of extract from trigonelline-rich plant have previously been reported [16]. A recent study has demonstrated the direct effects of trigonelline on COM crystals in kidney stone forming mechanism, including reduction of COM crystal size/number during crystallization, inhibition of COM crystal growth, and prevention of adherence of the crystals on cellular surfaces [17].
Renal epithelial cells form a monolayer to line along renal tubules. The epithelial cell sheet acts as a barrier for regulating fluid and electrolyte homeostasis as well as urine production. Defects in renal epithelial cells contribute to several kidney diseases, including chronic kidney disease (CKD) [18], renal fibrosis [19], diabetic nephropathy [20], and kidney stone disease [21]. Injury of the renal epithelial cell sheet can be induced by several factors, e.g., physical forces [22], drugs [23], high-glucose condition [24], chemicals [25], and crystals [26], [27]. During the past decade, the use of bioactive compounds for relieving and protecting renal epithelial cell injury has gained wide attention [28], [29], [30]. However, precise protective cellular mechanisms and response of renal cells to trigonelline remain hazy.
Therefore, we aimed to explore cellular mechanisms underlying the anti-lithiatic effects of trigonelline. Renal epithelial cells were treated with physiologic (subtoxic) concentration of trigonelline and subjected to quantitative proteomics to assess altered cellular proteome in response to trigonelline. Functional enrichment and reactome pathway analyses were performed to determine relevant biological pathways associated with the trigonelline-induced altered proteins. Finally, corresponding functional assays were performed to validate such relevant biological pathways related to the protective cellular mechanisms and the anti-lithiatic effects of trigonelline.
2. Materials and Methods
2.1. Cultivation of renal cells
MDCK (Madin-Darby canine kidney) cells (ATCC; Manassas, VA) were grown in a growth medium containing DMEM (Gibco; Grand Island, NY), antibiotics (penicillin G at 60 U/ml and streptomycin at 60 μg/ml) (Sigma-Aldrich; St. Louis, MO) and 10% fetal bovine serum. The cells were maintained in a CO2 incubator (Thermo Fisher Scientific; Marietta, OH) at 37 °C with 5% CO2 and relative humidity > 95%.
2.2. Trigonelline treatment
Approximately 1 × 106 cells were suspended in 2 ml growth medium and seeded into 6-well culture plate (Corning Inc.; Corning, NY). After 24-h incubation, the growth medium was refreshed and added with 1, 10 or 100 µM trigonelline (Sigma-Aldrich). The cells were further incubated with or without trigonelline for 24-h and then subjected to subsequent experiments as detailed below.
2.3. Cell death evaluation
Trypan blue exclusion assay was performed to select optimal concentration of trigonelline for subsequent experiments. The optimal concentration was set as the maximum that did not significantly induce cell death. After 24-h incubation with/without trigonelline, cell morphology was examined under an Eclipse Ti-S phase-contrast inverted microscope (Nikon; Tokyo, Japan). Floating and adherent cells were harvested and stained with 0.4% trypan blue solution (Gibco). Using a hemacytometer, the stained dead cells and unstained viable cells were counted and percentage of cell death was calculated as follows.
| Cell death (%) = (No. of dead cells / No. of viable and dead cells) × 100% |
2.4. In-solution tryptic digestion, nanoLC-ESI-Qq-TOF MS/MS analysis, and label-free quantitative proteomics
After 24-h incubation with/without 100 µM trigonelline, cellular proteins were extracted using SDT lysis buffer (4% SDS, 100 mM DTT, and 100 mM Tris-HCl; pH 7.6). Protein concentrations were measured using Bio-Rad protein assay (Bio-Rad; Milano, Italy) based on Bradford’s method. Equal amount (30 µg) of proteins from each sample was used for in-solution tryptic digestion as described previously [31], [32]. The digested peptides were then analyzed by nanoLC-ESI-LTQ-Orbitrap MS/MS as previously reported [33], [34]. The MS raw files (.d) were analyzed using MaxQuant software package (version 1.6.2.6) (https://www.maxquant.org) with built-in Andromeda search engine as described elsewhere [35]. More details of tryptic digestion and mass spectrometric analyses are provided in Supplementary Methods.
For quantitative analysis, label‐free quantification (LFQ) option was enabled with a minimum ratio count of 2. The LFQ intensity (derived from three biological replicates per group and technical triplicate per each biological sample) (generated according to the MaxLFQ algorithm [36]) was used as the quantified value. Peptides with less than five out of nine quantified values in each group were excluded from quantitative analysis. Multivariate statistical analysis was performed to determine the separation of the data derived from each group. The ropls R package [37] was employed to create orthogonal partial least squares discriminant analysis (OPLS-DA) model. The model quality was evaluated with R2X, R2Y (representing good fitness of the model) and Q2Y (representing predictability of the model). To avoid model overfitting, the OPLS-DA model was validated by implementing seven-fold cross validation and applying 200 random permutations. The variable influence on projection (VIP) values indicating the impact of the quantified proteins in the OPLS-DA model were computed. The protein quantification and univariate statistical analysis were carried out using unpaired Student’s t-test. The proteins with p values < 0.05 and VIP values > 1 were considered as significantly altered proteins.
2.5. Western blotting
After 24-h incubation with/without 100 µM trigonelline, cellular proteins were extracted with Laemmli’s buffer and protein concentrations were measured using Bio-Rad protein assay based on Bradford’s method. An equal amount (30 µg) of total proteins from each sample was subjected to Western blotting as described previously [38], [39]. See more details in Supplementary Methods.
2.6. Functional enrichment and reactome pathway analyses
ClueGO plugin (version 2.5.9) (https://apps.cytoscape.org/apps/cluego) and Cytoscape software (version 3.9.1) (https://cytoscape.org/) were used to acquire functional enrichment and reactome biological pathways of all significantly altered proteins induced by trigonelline based on the biological process branch of Gene Ontology (GO).
2.7. Calcium oxalate monohydrate (COM) crystal treatment
COM crystals were produced using the method published previously [40], [41]. After 24-h pretreatment with/without 100 µM trigonelline, the cells were incubated with COM crystals (100 µg crystal/ml medium) for further 24-h. The cells were then subjected to quantitative analysis of intracellular reactive oxygen species (ROS), cell cycle analysis, transepithelial electrical resistance (TEER) measurement, evaluation of zonula occludens-1 (ZO-1) level, and measurements of cell spindle index, epithelial marker (E-cadherin) and mesenchymal marker (vimentin) as detailed below. In parallel, the cells without trigonelline and COM crystal treatments served as the control.
2.8. Quantitative analysis of intracellular ROS
After 24-h pretreatment with/without 100 µM trigonelline followed by 24-h incubation with COM crystals, the cells were subjected to quantitative analysis of intracellular ROS level using DCFH-DA (dichlorodihydrofluorescein diacetate) assay and flow cytometry as previously described [42], [43]. Briefly, the cells were detached by trypsinization and resuspended in the growth medium followed by 20-min incubation with 5 μM DCFH-DA (Invitrogen-Molecular Probes; Burlington, Canada) at 37 °C in the dark. A flow cytometer (BD Accuri C6) equipped with Accuri C6 software (BD Biosciences; San Jose, CA) was employed to quantify the DCFH-DA-positive cells from at least 10,000 acquisitions in each sample.
2.9. Cell cycle analysis
After 24-h pretreatment with/without 100 µM trigonelline followed by 24-h incubation with COM crystals, the cells were subjected to flow cytometric analysis of cell cycle distribution as described previously [44], [45]. Briefly, the adherent cells were detached by trypsinization and incubated with ice-cold 70% ethanol for 30-min. After washing with ice-cold PBS, the cells were incubated with 100 μg/ml RNase A (Sigma-Aldrich) in PBS on ice for 30-min. The cells were further stained with propidium iodide (BD Biosciences) at 25 °C for 10-min in the dark. BD Accuri 6 flow cytometer equipped with Accuri C6 software (BD Biosciences) was employed to quantify the cells from at least 10,000 acquisitions in each sample. Cell cycle distribution was then analyzed according to DNA contents of the stained cells.
2.10. TEER measurement
TEER measurement was performed as previously described [46], [47]. Briefly, polyethylene culture inserts (surface area 1.12 cm2) of the 12-mm Transwell plate (0.4-µm pore size) (Corning) were pre-coated with collagen type IV (Sigma-Aldrich). The cells resuspended in the growth medium were seeded onto the upper chamber, whereas the lower chamber was filled with the growth medium. The polarized MDCK cells were subjected to 24-h pretreatment with/without 100 µM trigonelline followed by 24-h incubation with COM crystals as described above. The cells were then subjected to TEER measurement using Millicell-ERS Voltohmmeter equipped with chopstick electrode (Millipore; Bedford, MA). The TEER values were measured from at least three different sites in each well. The cell-free wells filled only with the growth medium severed as the blank for background subtraction. TEER values were calculated as follows.
| TEER (Ω·cm2) = (Resistance sample – Resistance blank) (Ω)× culture area (cm2) |
2.11. Immunofluorescence staining
Immunofluorescence study was performed as described previously [48], [49]. Briefly, the cells were grown on cover slips and subjected to 24-h pretreatment with/without 100 µM trigonelline followed by 24-h incubation with COM crystals as described above. The cells were then rinsed with PBS containing 0.1 mM CaCl2 and 1 mM MgCl2 and fixed and permeabilized with 4% paraformaldehyde and 0.1% TritonX-100, respectively (15-min each). After extensive wash, they were incubated with 5% bovine serum albumin (BSA) in PBS and then with mouse monoclonal anti-ZO-1 antibody (Invitrogen) (1:100 in 1% BSA/PBS) at 4 °C overnight. After washing, the cells were incubated with Alexa Fluor 488-conjugated corresponding secondary antibody (Invitrogen) (1:10,000 in 1% BSA/PBS) mixed with Hoechst dye (Invitrogen) (1:1000) at 37 °C for 1-h. After washing and mounting on glass slides, the cells were imaged under Eclipse 80i fluorescence microscope equipped with NIS-Elements D software (version 4.11) (Nikon). Fluorescence intensities were quantified from at least 100 cells in ≥ 10 random high-power fields (HPFs) per each sample. Their spectral data were also examined to determine distribution and localization of the protein.
2.12. Cell spindle index measurement
After 24-h pretreatment with/without 100 µM trigonelline followed by 24-h incubation with COM crystals, cell images were captured using an Eclipse Ti-S phase-contrast inverted microscope equipped with NIS-Elements D software (version 4.11) (Nikon). Using this software, lengths and widths of at least 100 cells in ≥ 10 random HPFs per sample were measured to calculate spindle index [50], [51] as follows.
| Spindle index = Cell length / Cell width |
2.13. Statistical analysis
The data are reported as mean ± standard deviation (SD) (unless stated otherwise) of measurements obtained from three separate experiments using independent biological samples. Unpaired Student’s t-test was performed to determine difference between two groups, whereas one-way ANOVA with Tukey’s post-hoc test was used for comparing more than two groups of samples. P value < 0.05 was set as the significant threshold.
3. Results
3.1. Effects of trigonelline on the morphology and viability of MDCK cells
The cells were incubated with trigonelline at 1, 10 or 100 µM for 24-h. Cell morphology was examined and dead cells were quantified. The data showed neither obvious morphological changes (Fig. 1A) nor significant increase in cell death (Fig. 1B) induced by any of these concentrations of trigonelline as compared with the untreated control. Therefore, trigonelline at 100 µM was chosen for all following experiments.
Fig. 1.
Optimal concentration for trigonelline treatment. MDCK cells were treated with trigonelline (TRIG) at 1, 10 or 100 µM for 24-h. (A): Cell morphology was examined under an Eclipse Ti-S phase-contrast inverted microscope (Nikon). (B): Percentage of cell death was quantified. Each dot represents each data value, whereas the bar indicates mean ± SD. The data were acquired from three separate experiments using independent biological samples. No significant differences among groups were detected.
3.2. Trigonelline induced changes in levels of several proteins in MDCK cells
From a total of 1006 and 1011 proteins identified from control and trigonelline-treated MDCK cells, respectively, MaxQuant-based quantitative proteomics using nanoLC-ESI-Qq-TOF MS/MS revealed 62 differentially expressed proteins between groups. These include 23 upregulated and 39 downregulated proteins induced by trigonelline (Fig. 2A and Table 1). Discrimination of the sample groups by orthogonal partial least squares discriminant analysis (OPLS-DA) showed the clear separation of the control and trigonelline-treated groups (Fig. 3A). Additionally, permutations could significantly confirm the generated OPLS-DA model (Fig. 3B).
Fig. 2.
Label-free quantitative proteomics and confirmation by Western blotting. After 24-h incubation with/without 100 µM trigonelline (TRIG), cellular proteins were extracted and subjected to in-solution tryptic digestion, nanoLC-ESI-Qq-TOF MS/MS analysis, and label-free quantitative proteomics. (A): A Venn diagram illustrating numbers of total and differentially expressed proteins identified from control and trigonelline-treated cells. (B): Western blotting to confirm the increased level of annexin A2 and decreased level of β-actin induced by trigonelline. GAPDH served as the loading control. (C): Band intensity of each protein was quantified and normalized by that of GAPDH. Each dot represents each data value, whereas the bar indicates mean ± SD. The data were acquired from three separate experiments using independent biological samples. Significant P values are labelled.
Table 1.
Summary of proteins with significantly altered levels in MDCK cells induced by trigonelline.
| Protein name | Swiss-Prot ID | Gene symbol | MS/MS identification score | %Cov | No. of distinct/ total matched peptides | MW (kDa) |
LFQ intensity (×105a.u.) (Mean ± SEM) |
Ratio (Trigonelline/Control) | Pvalue | VIP value | |
|---|---|---|---|---|---|---|---|---|---|---|---|
| Control | Trigonelline | ||||||||||
| 10 kDa heat shock protein, mitochondrial | P61604 | HSPE1 | 37.63 | 73.5 | 9/9 | 10.93 | 57.39 ± 1.42 | 53.05 ± 0.88 | 0.9244 | 0.0192 | 2.0135 |
| 14–3–3 protein eta | P68511 | YWHAH | 33.75 | 39.4 | 6/10 | 28.21 | 29.61 ± 0.75 | 26.77 ± 0.88 | 0.9042 | 0.0253 | 1.9516 |
| 26 S proteasome non-ATPase regulatory subunit 1 | Q99460 | PSMD1 | 15.16 | 7.7 | 5/5 | 105.84 | 8.65 ± 0.55 | 5.66 ± 1.18 | 0.6547 | 0.0353 | 1.7682 |
| 40 S ribosomal protein S12 | Q76I81 | RPS12 | 151.96 | 74.2 | 9/9 | 14.52 | 72.85 ± 1.68 | 66.29 ± 2.43 | 0.9100 | 0.0411 | 1.7912 |
| 40 S ribosomal protein S28 | Q6QAT1 | RPS28 | 19.55 | 56.5 | 5/5 | 7.84 | 11.45 ± 1.67 | 17.83 ± 1.68 | 1.5573 | 0.0158 | 2.0441 |
| 40 S ribosomal protein S29 | Q6QAP6 | RPS29 | 3.16 | 26.8 | 2/2 | 6.60 | 2.12 ± 1.40 | 7.53 ± 1.90 | 3.5512 | 0.0356 | 1.8017 |
| 40 S ribosomal protein S4 | P79103 | RPS4 | 109.74 | 45.2 | 14/14 | 29.60 | 57.29 ± 1.19 | 65.68 ± 2.32 | 1.1465 | 0.0054 | 2.2702 |
| 40 S ribosomal protein S8 | Q5E958 | RPS8 | 105.71 | 48.6 | 9/9 | 24.21 | 58.31 ± 1.05 | 61.39 ± 0.89 | 1.0529 | 0.0397 | 1.8091 |
| 40 S ribosomal protein S9 | Q6ZWN5 | RPS9 | 30.77 | 46.9 | 10/10 | 22.59 | 62.65 ± 1.85 | 68.37 ± 1.20 | 1.0913 | 0.0198 | 1.9884 |
| 60 S ribosomal protein L19 | Q3T0W9 | RPL19 | 100.21 | 21.9 | 6/6 | 23.47 | 44.20 ± 0.87 | 40.66 ± 1.09 | 0.9200 | 0.0216 | 1.9446 |
| 60 S ribosomal protein L35 | Q3MHM7 | RPL35 | 21.10 | 29.3 | 4/4 | 14.57 | 12.95 ± 1.47 | 21.98 ± 2.16 | 1.6974 | 0.0032 | 2.3903 |
| Actin, cytoplasmic 2 | P63261 | ACTG1 | 323.31 | 76.8 | 1/29 | 41.79 | 770.37 ± 11.22 | 692.01 ± 13.92 | 0.8983 | 0.0005 | 2.6759 |
| Actin-related protein 2 | Q5M7U6 | ACTR2 | 23.36 | 15.0 | 4/4 | 44.73 | 12.98 ± 0.37 | 11.95 ± 0.29 | 0.9209 | 0.0442 | 1.7199 |
| Actin-related protein 2/3 complex subunit 4 | Q148J6 | ARPC4 | 7.78 | 16.1 | 3/3 | 19.67 | 9.28 ± 0.25 | 10.32 ± 0.21 | 1.1112 | 0.0069 | 2.2276 |
| ADP/ATP translocase 3 | P32007 | SLC25A6 | 22.51 | 30.9 | 4/11 | 32.88 | 32.35 ± 1.32 | 36.05 ± 1.07 | 1.1143 | 0.0442 | 1.7542 |
| Alpha-actinin-1 | Q7TPR4 | ACTN1 | 16.89 | 18.6 | 5/14 | 103.07 | 10.34 ± 0.49 | 12.39 ± 0.57 | 1.1978 | 0.0157 | 2.0352 |
| Annexin A2 | Q6TEQ7 | ANXA2 | 323.31 | 69.3 | 26/26 | 38.65 | 285.46 ± 4.95 | 321.17 ± 8.84 | 1.1251 | 0.0028 | 2.4381 |
| Annexin A7 | P20073 | ANXA7 | 4.06 | 7.4 | 3/3 | 52.74 | 2.25 ± 0.89 | 0.00 ± 0.00 | 0.0000 | 0.0227 | 1.9381 |
| Asparagine synthetase [glutamine-hydrolyzing] | Q1LZA3 | ASNS | 26.46 | 21.4 | 1/9 | 64.22 | 1.53 ± 0.64 | 4.35 ± 0.99 | 2.8391 | 0.0300 | 1.8745 |
| Asparagine--tRNA ligase, cytoplasmic | O43776 | NARS1 | 2.88 | 5.3 | 2/2 | 62.94 | 4.28 ± 0.16 | 1.34 ± 0.67 | 0.3126 | 0.0006 | 2.6651 |
| ATP synthase subunit alpha, mitochondrial | P19483 | ATP5F1A | 253.06 | 48.1 | 1/23 | 59.72 | 109.94 ± 2.12 | 125.39 ± 2.47 | 1.1405 | 0.0002 | 2.7768 |
| ATP-citrate synthase | Q32PF2 | ACLY | 36.89 | 12.5 | 1/10 | 119.79 | 20.64 ± 0.48 | 19.33 ± 0.32 | 0.9363 | 0.0366 | 1.8374 |
| Basic leucine zipper and W2 domain-containing protein 1 | Q9CQC6 | BZW1 | 14.40 | 17.7 | 6/6 | 48.04 | 0.00 ± 0.00 | 2.89 ± 1.15 | #DIV/0! | 0.0229 | 1.9511 |
| Cell division control protein 42 homolog | Q8CFN2 | CDC42 | 12.96 | 31.4 | 4/5 | 21.26 | 10.67 ± 0.80 | 15.83 ± 1.70 | 1.4845 | 0.0142 | 2.0710 |
| Cytosolic non-specific dipeptidase | Q3ZC84 | CNDP2 | 3.58 | 4.0 | 1/2 | 52.66 | 1.76 ± 0.56 | 0.00 ± 0.00 | 0.0000 | 0.0061 | 2.2440 |
| DNA replication licensing factor MCM5 | P33992 | MCM5 | 6.57 | 8.9 | 5/5 | 82.29 | 4.77 ± 1.22 | 1.39 ± 0.92 | 0.2920 | 0.0421 | 1.7688 |
| DNA replication licensing factor MCM6 | Q14566 | MCM6 | 7.86 | 6.0 | 4/4 | 92.89 | 7.63 ± 0.19 | 3.84 ± 1.22 | 0.5033 | 0.0072 | 2.2345 |
| Eukaryotic translation initiation factor 3 subunit I | Q5E966 | EIF3I | 7.53 | 29.8 | 5/5 | 36.47 | 8.94 ± 0.21 | 4.00 ± 1.58 | 0.4476 | 0.0070 | 2.2145 |
| Far upstream element-binding protein 2 | Q92945 | KHSRP | 22.70 | 17.0 | 10/10 | 73.11 | 22.09 ± 0.32 | 20.40 ± 0.43 | 0.9236 | 0.0058 | 2.2686 |
| Glucose-6-phosphate isomerase | P06744 | GPI | 11.22 | 5.0 | 2/2 | 63.15 | 2.81 ± 0.90 | 5.78 ± 0.40 | 2.0542 | 0.0081 | 2.2118 |
| GTP-binding nuclear protein Ran | Q3T054 | RAN | 53.60 | 33.3 | 8/8 | 24.42 | 74.35 ± 0.81 | 68.47 ± 1.15 | 0.9209 | 0.0007 | 2.6611 |
| Heterogeneous nuclear ribonucleoprotein F | Q5E9J1 | HNRNPF | 44.38 | 20.5 | 5/7 | 45.69 | 18.54 ± 0.94 | 15.56 ± 0.69 | 0.8393 | 0.0218 | 1.9753 |
| Heterogeneous nuclear ribonucleoprotein L | Q8R081 | HNRNPL | 61.14 | 25.6 | 11/11 | 63.96 | 30.63 ± 0.73 | 24.65 ± 1.02 | 0.8049 | 0.0002 | 2.7787 |
| Heterogeneous nuclear ribonucleoproteins A2/B1 | O88569 | HNRNPA2B1 | 252.84 | 41.1 | 13/13 | 37.40 | 101.20 ± 2.34 | 92.46 ± 3.01 | 0.9136 | 0.0356 | 1.7923 |
| Hexokinase-2 | P52789 | HK2 | 4.80 | 3.2 | 2/2 | 102.38 | 4.64 ± 0.59 | 2.21 ± 0.88 | 0.4773 | 0.0353 | 1.8302 |
| Lamin-B1 | P14733 | LMNB1 | 198.28 | 35.7 | 4/26 | 66.79 | 113.41 ± 2.73 | 105.36 ± 2.08 | 0.9290 | 0.0320 | 1.8425 |
| Matrin-3 | P43243 | MATR3 | 40.77 | 11.8 | 1/6 | 94.62 | 14.27 ± 0.48 | 12.76 ± 0.37 | 0.8942 | 0.0240 | 1.9348 |
| Mitochondrial import inner membrane translocase subunit Tim13 | Q9Y5L4 | TIMM13 | 4.40 | 25.3 | 2/2 | 10.50 | 2.27 ± 0.58 | 0.42 ± 0.42 | 0.1849 | 0.0198 | 1.9990 |
| Myosin-9 | Q8VDD5 | MYH9 | 323.31 | 26.9 | 38/45 | 226.37 | 108.55 ± 2.38 | 116.03 ± 1.39 | 1.0689 | 0.0152 | 2.0276 |
| NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13 | Q95KV7 | NDUFA13 | 4.11 | 13.9 | 2/2 | 16.67 | 3.36 ± 0.67 | 1.12 ± 0.56 | 0.3339 | 0.0209 | 1.9603 |
| Non-POU domain-containing octamer-binding protein | Q15233 | NONO | 76.26 | 33.1 | 11/11 | 54.23 | 41.76 ± 0.86 | 38.28 ± 1.31 | 0.9167 | 0.0415 | 1.7688 |
| Non-specific lipid-transfer protein | P32020 | SCP2 | 172.41 | 12.4 | 1/7 | 59.13 | 26.36 ± 1.53 | 33.89 ± 2.34 | 1.2859 | 0.0159 | 2.0250 |
| Peptidyl-prolyl cis-trans isomerase B | P23284 | PPIB | 100.57 | 25.9 | 5/5 | 23.74 | 32.18 ± 0.95 | 35.66 ± 1.03 | 1.1082 | 0.0243 | 1.9118 |
| Peptidyl-tRNA hydrolase 2, mitochondrial | Q3ZBL5 | PTRH2 | 11.39 | 18.4 | 3/3 | 19.29 | 0.00 ± 0.00 | 2.07 ± 0.69 | #DIV/0! | 0.0086 | 2.1839 |
| Poly(U)-binding-splicing factor PUF60 | Q2HJG2 | PUF60 | 22.37 | 14.7 | 4/4 | 57.09 | 6.28 ± 0.35 | 4.75 ± 0.63 | 0.7558 | 0.0493 | 1.7012 |
| Probable ATP-dependent RNA helicase DDX17 | Q501J6 | DDX17 | 56.20 | 22.9 | 9/13 | 72.40 | 19.53 ± 0.43 | 18.00 ± 0.40 | 0.9212 | 0.0184 | 2.0230 |
| Proteasome activator complex subunit 3 | P61290 | PSME3 | 15.05 | 28.3 | 6/6 | 29.51 | 8.71 ± 0.29 | 7.70 ± 0.26 | 0.8835 | 0.0191 | 1.9863 |
| Proteasome subunit alpha type-2 | Q3T0Y5 | PSMA2 | 2.95 | 14.1 | 2/2 | 25.90 | 0.00 ± 0.00 | 4.30 ± 1.38 | #DIV/0! | 0.0067 | 2.2294 |
| Proteasome subunit beta type-6 | P28072 | PSMB6 | 8.34 | 17.6 | 2/4 | 25.36 | 18.81 ± 0.91 | 15.81 ± 0.62 | 0.8404 | 0.0150 | 2.0664 |
| Protein arginine N-methyltransferase 1 | Q63009 | PRMT1 | 22.63 | 22.9 | 6/6 | 40.52 | 17.60 ± 0.47 | 15.83 ± 0.53 | 0.8994 | 0.0240 | 1.9421 |
| Protein PBDC1 | Q9BVG4 | PBDC1 | 4.99 | 9.4 | 2/2 | 26.06 | 9.02 ± 0.42 | 5.34 ± 1.39 | 0.5921 | 0.0222 | 1.9862 |
| Protein transport protein Sec23B | Q9D662 | SEC23B | 2.20 | 3.8 | 2/2 | 86.44 | 0.61 ± 0.40 | 2.11 ± 0.54 | 3.4601 | 0.0408 | 1.7612 |
| Prothymosin alpha | P06454 | PTMA | 38.92 | 34.2 | 4/4 | 12.20 | 14.83 ± 2.11 | 6.24 ± 3.17 | 0.4206 | 0.0383 | 1.8681 |
| Radixin | P26043 | RDX | 27.73 | 20.8 | 3/11 | 68.54 | 4.07 ± 0.53 | 1.87 ± 0.74 | 0.4585 | 0.0283 | 1.9012 |
| Ras-related protein Rab-1B | Q9H0U4 | RAB1B | 70.79 | 54.2 | 5/11 | 22.17 | 28.83 ± 1.14 | 33.33 ± 1.01 | 1.1558 | 0.0095 | 2.1429 |
| Retinal dehydrogenase 1 | P48644 | ALDH1A1 | 16.42 | 10.6 | 1/4 | 54.81 | 5.28 ± 0.70 | 2.05 ± 1.05 | 0.3887 | 0.0208 | 1.9586 |
| RuvB-like 1 | Q9Y265 | RUVBL1 | 33.88 | 37.3 | 13/13 | 50.23 | 26.12 ± 0.47 | 24.56 ± 0.50 | 0.9402 | 0.0376 | 1.8400 |
| Small nuclear ribonucleoprotein F | Q3T0Z8 | SNRPF | 5.98 | 24.4 | 2/2 | 9.73 | 5.80 ± 1.12 | 1.57 ± 1.04 | 0.2709 | 0.0138 | 2.0907 |
| Succinate dehydrogenase [ubiquinone] flavoprotein subunit, mitochondrial | Q8K2B3 | SDHA | 32.87 | 8.4 | 3/4 | 72.59 | 12.12 ± 0.22 | 11.04 ± 0.39 | 0.9105 | 0.0277 | 1.8591 |
| Synaptic vesicle membrane protein VAT-1 homolog | Q99536 | VAT1 | 10.02 | 13.0 | 4/4 | 41.92 | 3.19 ± 1.01 | 0.58 ± 0.58 | 0.1820 | 0.0402 | 1.7829 |
| Transcription intermediary factor 1-beta | Q13263 | TRIM28 | 16.61 | 9.8 | 6/6 | 88.55 | 12.39 ± 0.37 | 11.14 ± 0.44 | 0.8992 | 0.0457 | 1.7365 |
| Transmembrane emp24 domain-containing protein 9 | Q9BVK6 | TMED9 | 55.45 | 17.4 | 4/4 | 27.28 | 7.20 ± 0.18 | 8.19 ± 0.22 | 1.1372 | 0.0031 | 2.4035 |
%Cov = (No. of the identified amino acid residues/No. of amino acid residues in the protein sequence) × 100.
LFQ = label-free quantification.
SEM = standard error of mean.
#DIV/0! = divided by zero.
VIP value = variable influence on projection value (obtained from the OPLS-DA model generated using ropls R package).
Fig. 3.
Discrimination of the sample groups by orthogonal partial least squares discriminant analysis (OPLS-DA). (A): Data distribution and the OPLS-DA scores calculated using the ropls R package to discriminate the control (C, red) and trigonelline-treated (T, blue) groups. The OPLS-DA score plot displays clear separation of the control and trigonelline-treated groups. (B): The 200 random permutations could significantly confirm the generated OPLS-DA model.
From these, upregulation of annexin A2 and downregulation of β-actin were randomly selected to be confirmed by Western blotting. The results confirmed significantly increased level of annexin A2 and significantly decreased level of β-actin in trigonelline-treated cells, consistent with the mass spectrometric data (Fig. 2B and C).
3.3. Biological processes/pathways involved by the trigonelline-induced significantly altered proteins
To predict relevant functions of all these significantly altered proteins induced by trigonelline, their gene symbols were submitted for functional enrichment and reactome pathway analyses using the ClueGO plug-in and Cytoscape software. Functional enrichment analysis showed that the biological processes involved by these trigonelline-induced significantly altered proteins included cellular response to stress, cell cycle and cell polarity (Fig. 4A). Reactome pathway analysis also revealed that cellular response to stress, cell cycle and cell polarity were the three main biological pathways involved by these trigonelline-induced significantly altered proteins (Fig. 4B). Validation of these predictions was then performed using COM crystal as the stress/disease inducer, whereas trigonelline served as the protector/rescuer.
Fig. 4.
Functional enrichment and reactome pathway analyses of significantly altered proteins. All significantly altered proteins induced by trigonelline were subjected to functional enrichment analysis (A) and reactome pathway analysis (B) using the ClueGO plug-in and Cytoscape software. The GO terms of biological processes and individual proteins involved in each biological pathway are labelled.
3.4. Trigonelline prevented COM crystal-induced intracellular ROS overproduction
After 24-h pretreatment with/without 100 µM trigonelline followed by 24-h incubation with COM crystals, intracellular ROS level was measured using DCFH-DA assay combined with flow cytometry. Fig. 5A shows histogram of cell number (y-axis) and fluorescence intensity (x-axis) after DCFH-DA probing, whereas Fig. 5B demonstrates the quantitative analysis of the data shown in Fig. 5A. The analysis revealed that the intracellular ROS level was significantly elevated by COM crystals compared with the untreated control (Fig. 5). However, pretreatment with trigonelline completely prevented such increase of intracellular ROS level induced by COM crystals (Fig. 5).
Fig. 5.
Trigonelline prevented COM crystal-induced intracellular ROS overproduction. After 24-h pretreatment with/without 100 µM trigonelline (TRIG) followed by 24-h incubation with COM crystals, intracellular ROS level was measured using DCFH-DA assay and flow cytometry. (A): Histogram of the cell number and fluorescence intensity. (B): Percentage of the DCFH-DA-positive cells were quantified from at least 10,000 acquisitions in each sample. Each dot represents each data value, whereas the bar indicates mean ± SD. The data were acquired from three separate experiments using independent biological samples. Significant P values are labelled.
3.5. Trigonelline prevented COM crystal-induced cell cycle shift from G0/G1 to G2/M phase
After 24-h pretreatment with/without 100 µM trigonelline followed by 24-h incubation with COM crystals, DNA contents were analyzed by propidium iodide staining followed by flow cytometry to determine cell cycle distribution (Fig. 6A). Quantitative analysis of the cells in individual cycles showed that COM crystals caused significant decrease in cell population at G0/G1 phase and increase in cell population at G2/M phase, indicating cell cycle shift from G0/G1 to G2/M phase, as compared with the untreated control (Fig. 6B). However, trigonelline pretreatment successfully prevented such cell cycle shift induced by COM crystals (Fig. 6).
Fig. 6.
Trigonelline prevented COM crystal-induced cell cycle shift from G0/G1 to G2/M phase. After 24-h pretreatment with/without 100 µM trigonelline (TRIG) followed by 24-h incubation with COM crystals, DNA contents were analyzed by propidium iodide staining followed by flow cytometry to determine cell cycle distribution. (A): Histogram of the cell number and fluorescence intensity of each phase of the cell cycle. (B): Distribution of the cell population in G0/G1, S and G2/M phases of the cell cycle were quantified from at least 10,000 acquisitions in each sample. Each dot represents each data value, whereas the bar indicates mean ± SD. The data were acquired from three separate experiments using independent biological samples. Significant P values are labelled.
3.6. Trigonelline prevented COM crystal-induced tight junction disruption
After 24-h pretreatment with/without 100 µM trigonelline followed by 24-h incubation with COM crystals, transepithelial electrical resistance (TEER) was measured using Millicell-ERS Voltohmmeter, and expression of a tight junction protein (ZO-1) was examined by immunofluorescence staining and Western blotting. The data showed that COM crystals caused significant reduction of TEER (Fig. 7A). Immunofluorescence study revealed that COM crystals significantly suppressed expression level of ZO-1 (Fig. 7B). Moreover, spectral intensity data obviously showed the COM crystal-induced tight junction disruption at the cell borders (Fig. 7C). Western blotting confirmed the reduced level of ZO-1 in the COM-treated cells (Fig. 7D and E). Such markers for tight junction disruption induced by COM crystals were completely preserved at their basal levels by trigonelline pretreatment (Fig. 7).
Fig. 7.
Trigonelline prevented COM crystal-induced tight junction disruption. After 24-h pretreatment with/without 100 µM trigonelline (TRIG) followed by 24-h incubation with COM crystals, TEER and ZO-1 expression were measured. (A): TEER was measured using Millicell-ERS Voltohmmeter. (B): Immunofluorescence staining of ZO-1. (C): Spectral intensity of ZO-1 across the cells (from the area indicated by the red arrow). a.u. = arbitrary unit. (D): Western blotting of ZO-1 and GAPDH (loading control). (E): Band intensity of ZO-1 was quantified and normalized by that of GAPDH. Each dot represents each data value, whereas the bar indicates mean ± SD. The data were acquired from three separate experiments using independent biological samples. Significant P values are labelled.
3.7. Trigonelline prevented COM crystal-induced epithelial-mesenchymal transition (EMT)
After 24-h pretreatment with/without 100 µM trigonelline followed by 24-h incubation with COM crystals, EMT markers (including spindle index, E-cadherin and vimentin) were examined. The analyses revealed that COM crystals caused cell elongation (Fig. 8A), increased spindle index (Fig. 8B) and expression of vimentin (a mesenchymal marker) (Fig. 8C and D), but decreased expression of E-cadherin (an epithelial marker) (Fig. 8C and D), indicating the COM crystal-induced EMT process in renal epithelial cells. All of these EMT markers were successfully preserved by trigonelline pretreatment (Fig. 8).
Fig. 8.
Trigonelline prevented COM crystal-induced EMT. After 24-h pretreatment with/without 100 µM trigonelline (TRIG) followed by 24-h incubation with COM crystals, EMT markers were evaluated. (A): Cell morphology. (B): Spindle index was measured from at least 100 cells in ≥ 10 random HPFs per sample. (C): Western blotting of E-cadherin (epithelial marker) and vimentin (mesenchymal marker), whereas GAPDH served as the loading control. (D): Band intensity of each protein was quantified and normalized by that of GAPDH. Each dot represents each data value, whereas the bar indicates mean ± SD. The data were acquired from three separate experiments using independent biological samples. Significant P values are labelled.
4. Discussion
This study employed label-free quantitative proteomics approach to explore the response of renal cells to trigonelline. The range of trigonelline concentrations tested (1–100 µM) was chosen based on its physiologic range observed in humans after coffee consumption [8], [52], [53]. This range of trigonelline concentrations has also been used in several previous in vitro studies [54], [55], [56], [57]. Cell death analysis, which has been widely used for determination of severe cytotoxic effects from various chemicals, compounds and drugs [58], [59], [60], was applied to address the potential severe cytotoxic effect of trigonelline in the present study. Morphological examination and cell death analysis revealed that trigonelline at 1, 10 and 100 µM had no significant effects on cell morphology and death, consistent with the data reported in other studies indicating that trigonelline at 100 µM or lower concentrations does not cause severe cytotoxicity and significant increase in cell death in mesangial cells, hepatocytes and cardiocytes [61], [62], [63]. Therefore, its concentration at 100 µM was used for all following experiments to gain the maximal effects of this compound while its cytotoxic effects, if any, were minimal.
MaxQuant-based quantitative proteomics using nanoLC-ESI-Qq-TOF MS/MS identified 62 significantly altered proteins induced by trigonelline from over a thousand of proteins identified in renal cells. Quantitative proteomics generally provides a large variable dataset that reflects response of multiple proteins upon stimuli. Multivariate statistical analysis is thus commonly applied to find the meaning of such large variable dataset and to discriminate groups of the samples [64], [65]. The OPLS-DA, one of the multivariate statistical methods, is widely applied in several proteomics [66], [67], [68], [69] and metabolomics [70], [71], [72], [73] studies to discriminate the sample groups. Moreover, the VIP values greater than 1 are widely used to indicate the impact of the quantified proteins in the OPLS-DA model in many proteomics and metabolomics studies [70], [74], [75], [76]. In our study, the VIP values of all significantly altered proteins were > 1 (see Table 1), strengthening their impact on the OPLS-DA model to discriminate the trigonelline-treated group from the control. As mentioned above, the concentration of trigonelline used herein was at physiologic/subtoxic level. Therefore, it was not surprising that the number of significantly altered proteins was relatively small and the degree of changes (in term of fold-change) was relatively low as compared with other studies on chemicals-induced changes in cellular proteome (note that many of these studies do not address the cytotoxic effects from the tested chemicals). We therefore, used the VIP values > 1 (instead of fold-change) as another threshold for determining significant differences in addition to statistical P values. Among the significant differences identified, changes in levels of annexin A2 and β-actin induced by trigonelline were selected to be confirmed by Western blotting based on their highest MS/MS identification score at 323.31 (see Table 1) and the availability of their primary antibodies in our laboratory at the time of investigations. Functional prediction by GO biological process enrichment and reactome pathway analyses consistently guided that these trigonelline-induced altered proteins were involved in cellular response to stress, cell cycle and cell polarity.
According to many lines of evidence, trigonelline has beneficial effects on prevention of several diseases [11], [77]. Our recent study has also reported the direct effects of trigonelline on COM crystals to inhibit kidney stone formation [17]. However, cellular mechanisms underlying its anti-lithiatic effects remain largely unknown. Functional validation of the data predicted from enrichment and reactome analyses, therefore, focused on cellular mechanisms responsible for the anti-lithiatic effects of trigonelline.
Recent in vivo studies have found renal tissue injury, accompanied with ROS overproduction and oxidative stress, in ethylene glycol-induced nephrolithiatic rats [78], [79]. Additionally, recent in vitro studies have confirmed that COM crystals cause renal epithelial cell injury via induction of ROS overproduction and oxidative stress [28], [80], [81]. Our present study has shown the anti-oxidative effects of trigonelline, which completely prevented the increase of intracellular ROS induced by COM crystals. This finding is consistent with a previous study reporting that trigonelline can reduce intracellular ROS production induced by ultraviolet B (UVB) radiation in human foreskin fibroblasts [82]. Similarly, the anti-oxidative effects of trigonelline have also been reported to prevent UVB-induced oxidative damage of DNA contents in mice and human dermal fibroblasts [54]. Moreover, the anti-oxidative effects of trigonelline have been documented in several other in vivo studies using various disease models [13], [83], [84], [85]. These data underscore the anti-oxidative property of trigonelline that may be responsible for its anti-lithiatic effects.
DNA content analysis has revealed that COM crystals induced cell cycle shift from G0/G1 to G2/M phase, indicating cell cycle arrest at G2/M phase. In concordance, several recent studies have reported G2/M phase arrest caused by oxidative stress induced by various stimuli [86], [87], [88]. Additionally, G2/M phase arrest has been found in ischemia-induced proximal renal tubular cell injury [89]. The arrest at G2/M phase is also associated with development of renal fibrosis due to secretion of profibrogenic factors [89]. More importantly, cell cycle shift from G0/G1 to S and G2/M phases have been found in a recent study on scratch- and chemical-induced renal tubular cell injury [90]. These cells with cell cycle shift to S and G2/M phases induce COM crystal adhesion on their surfaces [90], thereby promoting the kidney stone mechanism. Our present study has shown the protective effects of trigonelline on COM-induced cell cycle shift from G0/G1 to G2/M phase, indicating that the anti-lithiatic effects of trigonelline may be mediated, at least in part, via preservation of cell cycle distribution of renal epithelial cells.
Cell polarity is a characteristic of epithelial cells that generally have two poles, apical and basolateral, to regulate and maintain cellular structure and functions. Similar to other epithelial cells, renal epithelial cells exhibit apico-basolateral poles that are separated by tight junctional protein complex [91]. This complex plays crucial roles in establishment and maintenance of renal cell polarity structure and functions. As such, loss or disruption of tight junction contributes to defective cell polarity and altered cellular functions. A recent study has reported that oxalate can induce renal epithelial barrier disruption via inhibition of Wnt/β-catenin signaling [92]. COM crystals also cause tight junction impairment through p38 MAPK signaling and intracellular ROS overproduction [93], [94]. Herein, we confirmed that COM crystals caused tight junction disruption as shown by the declines of TEER and ZO-1 expression. Interestingly, trigonelline could completely preserved tight junction structure and function. These data were consistent with other evidence reporting that bioactive compounds with anti-oxidant properties can alleviate stimuli-induced damages of tight junction complex [95], [96], [97].
Kidney stone disease also increases the risk for development of CKD [98]. Renal fibrosis is an important feature of CKD. One of the cellular mechanisms triggering renal fibrosis is EMT [99]. This transitional cellular process is characterized by loss of epithelial properties and gain of mesenchymal features of the cells [99]. A recent study has reported the EMT induction together with ROS accumulation in a mouse model of pulmonary fibrosis caused by high-glucose condition [100]. Herein, we found that COM induced EMT in renal epithelial cells as evidenced by the increased spindle index, decreased epithelial marker (E-cadherin) and increased mesenchymal marker (vimentin). Again, trigonelline successfully prevented these EMT features triggered by COM crystals. Our findings were consistent with the data reported in previous studies indicating that other natural bioactive compounds (i.e., epigallocatechin-3-gallate, caffeine, rhein and curcumin) with anti-oxidative and anti-inflammatory properties also exhibit the anti-fibrotic effects [50], [101], [102]. Additionally, a recent study on mesangial cells has revealed that trigonelline can reverse high-glucose-induced fibrosis by inhibition of Wnt signaling [55]. A previous study in neonatal diabetic rats has found that trigonelline can reduce renal fibrosis by suppressing oxidative stress [103]. Nevertheless, the evidence showing the advantages of trigonelline to prevent EMT process is limited. Only one recent study has shown that trigonelline can prevent high-oxalate-induced EMT progression [104]. These data implicate that, in addition to the protective mechanisms against kidney stone formation, trigonelline also prevents nephrolithiasis-associated CKD by preventing the EMT fibrotic mechanisms.
Finally, limitations of our present study should be also mentioned. Although a set of significantly altered proteins could lead to functional prediction followed by experimental validation, the number of significantly altered proteins was relatively small as compared with those reported in other studies. This was mainly due to the physiologic/subtoxic concentration of trigonelline selected and the mass spectrometric modality employed. Using the greater concentration of trigonelline and more sensitive mass spectrometric modality would enhance the identification of the significantly altered proteins in renal cells induced by trigonelline. However, the potential cytotoxicity of the greater concentration of trigonelline must be considered as it may interfere with the data interpretation. Additionally, the study was done with only a single time-point (24 h) for trigonelline treatment. Serial changes of the cellular proteome at various time-points would yield more in-depth insights and dynamicity of the trigonelline-induced changes in cellular proteome and function. Furthermore, we examined changes only in MDCK renal cells, whereas analyses on other renal cells (i.e., from other species and/or from different segments of nephron) would yield much greater dimension of the data. Finally, there are several other bioinformatic tools that can be used to enhance the interpretation and/or prediction of the proteome data. Having done so, the understanding of the biological effects of trigonelline in renal cells would be much clearer.
In summary, our present study has reported that trigonelline causes significant changes in levels of 62 (23 upregulated and 39 downregulated) proteins in MDCK cells. Functional enrichment and reactome pathway analyses reveal that these trigonelline-induced significantly altered proteins are related to stress response, cell cycle and cell polarity. Functional investigations confirm that trigonelline prevents COM crystal-induced renal cell deteriorations by inhibiting crystal-induced overproduction of intracellular ROS, cell cycle shift from G0/G1 to G2/M phase, tight junction disruption, and EMT in renal epithelial cells. These findings provide cellular mechanisms and convincing evidence for the renoprotective effects of trigonelline, particularly in kidney stone prevention.
CRediT authorship contribution statement
PPe, WB, PPu, SS, and VT designed research; PPe, WB, PPu, and SS performed experiments; PPe, WB, PPu, SS, and VT analyzed data; PPe and VT wrote the manuscript; All authors reviewed and approved the manuscript.
Declaration of Competing Interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Acknowledgements
This study is funded by National Research Council of Thailand (NRCT) and Mahidol University (grant no. N42A650371).
Footnotes
Supplementary data associated with this article can be found in the online version at doi:10.1016/j.csbj.2023.11.036.
Appendix A. Supplementary material
Supplementary material
.
References
- 1.Idris S., Mishra A., Khushtar M. Recent therapeutic interventions of fenugreek seed: a mechanistic approach. Drug Res (Stuttg) 2021;71:180–192. doi: 10.1055/a-1320-0479. [DOI] [PubMed] [Google Scholar]
- 2.Yadav U.C., Baquer N.Z. Pharmacological effects of Trigonella foenum-graecum l. In health and disease. Pharm Biol. 2014;52:243–254. doi: 10.3109/13880209.2013.826247. [DOI] [PubMed] [Google Scholar]
- 3.Honda M., Takezaki D., Tanaka M., Fukaya M., Goto M. Effect of roasting degree on major coffee compounds: a comparative study between coffee beans with and without supercritical co(2) decaffeination treatment. J Oleo Sci. 2022;71:1541–1550. doi: 10.5650/jos.ess22194. [DOI] [PubMed] [Google Scholar]
- 4.Dong R., Zhu M., Long Y., Yu Q., Li C., Xie J., et al. Exploring correlations between green coffee bean components and thermal contaminants in roasted coffee beans. Food Res Int. 2023;167 doi: 10.1016/j.foodres.2023.112700. [DOI] [PubMed] [Google Scholar]
- 5.Caprioli G., Cortese M., Maggi F., Minnetti C., Odello L., Sagratini G., et al. Quantification of caffeine, trigonelline and nicotinic acid in espresso coffee: The influence of espresso machines and coffee cultivars. Int J Food Sci Nutr. 2014;65:465–469. doi: 10.3109/09637486.2013.873890. [DOI] [PubMed] [Google Scholar]
- 6.Konstantinidis N., Franke H., Schwarz S., Lachenmeier D.W. Risk assessment of trigonelline in coffee and coffee by-products. Molecules. 2023;28:3460. doi: 10.3390/molecules28083460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Stennert A., Maier H.G. Trigonelline in coffee. Ii. Content of green, roasted and instant coffee. Z Leb Unters Forsch. 1994;199:198–200. doi: 10.1007/BF01193443. [DOI] [PubMed] [Google Scholar]
- 8.Lang R., Wahl A., Stark T., Hofmann T. Urinary n-methylpyridinium and trigonelline as candidate dietary biomarkers of coffee consumption. Mol Nutr Food Res. 2011;55:1613–1623. doi: 10.1002/mnfr.201000656. [DOI] [PubMed] [Google Scholar]
- 9.Guertin K.A., Moore S.C., Sampson J.N., Huang W.Y., Xiao Q., Stolzenberg-Solomon R.Z., et al. Metabolomics in nutritional epidemiology: Identifying metabolites associated with diet and quantifying their potential to uncover diet-disease relations in populations. Am J Clin Nutr. 2014;100:208–217. doi: 10.3945/ajcn.113.078758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Rothwell J.A., Madrid-Gambin F., Garcia-Aloy M., Andres-Lacueva C., Logue C., Gallagher A.M., et al. Biomarkers of intake for coffee, tea, and sweetened beverages. Genes Nutr. 2018;13:15. doi: 10.1186/s12263-018-0607-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Liang Y., Dai X., Cao Y., Wang X., Lu J., Xie L., et al. The neuroprotective and antidiabetic effects of trigonelline: a review of signaling pathways and molecular mechanisms. Biochimie. 2023;206:93–104. doi: 10.1016/j.biochi.2022.10.009. [DOI] [PubMed] [Google Scholar]
- 12.Farid M.M., Yang X., Kuboyama T., Tohda C. Trigonelline recovers memory function in alzheimer's disease model mice: evidence of brain penetration and target molecule. Sci Rep. 2020;10 doi: 10.1038/s41598-020-73514-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Fahanik-Babaei J., Baluchnejadmojarad T., Nikbakht F., Roghani M. Trigonelline protects hippocampus against intracerebral abeta(1-40) as a model of alzheimer's disease in the rat: insights into underlying mechanisms. Metab Brain Dis. 2019;34:191–201. doi: 10.1007/s11011-018-0338-8. [DOI] [PubMed] [Google Scholar]
- 14.Siener R., Herwig H., Rudy J., Schaefer R.M., Lossin P., Hesse A. Urinary stone composition in germany: results from 45,783 stone analyses. World J Urol. 2022;40:1813–1820. doi: 10.1007/s00345-022-04060-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Halinski A., Bhatti K.H., Boeri L., Cloutier J., Davidoff K., Elqady A., et al. Stone composition of renal stone formers from different global regions. Arch Ital Urol Androl. 2021;93:307–312. doi: 10.4081/aiua.2021.3.307. [DOI] [PubMed] [Google Scholar]
- 16.Laroubi A., Touhami M., Farouk L., Zrara I., Aboufatima R., Benharref A., et al. Prophylaxis effect of trigonella foenum graecum l. Seeds on renal stone formation in rats. Phytother Res. 2007;21:921–925. doi: 10.1002/ptr.2190. [DOI] [PubMed] [Google Scholar]
- 17.Peerapen P., Boonmark W., Thongboonkerd V. Trigonelline prevents kidney stone formation processes by inhibiting calcium oxalate crystallization, growth and crystal-cell adhesion, and downregulating crystal receptors. Biomed Pharm. 2022;149 doi: 10.1016/j.biopha.2022.112876. [DOI] [PubMed] [Google Scholar]
- 18.Liu B.C., Tang T.T., Lv L.L., Lan H.Y. Renal tubule injury: a driving force toward chronic kidney disease. Kidney Int. 2018;93:568–579. doi: 10.1016/j.kint.2017.09.033. [DOI] [PubMed] [Google Scholar]
- 19.Liu B.C., Tang T.T., Lv L.L. How tubular epithelial cell injury contributes to renal fibrosis. Adv Exp Med Biol. 2019;1165:233–252. doi: 10.1007/978-981-13-8871-2_11. [DOI] [PubMed] [Google Scholar]
- 20.Chang J., Yan J., Li X., Liu N., Zheng R., Zhong Y. Update on the mechanisms of tubular cell injury in diabetic kidney disease. Front Med (Lausanne) 2021;8 doi: 10.3389/fmed.2021.661076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Alelign T., Petros B. Kidney stone disease: an update on current concepts. Adv Urol. 2018;2018 doi: 10.1155/2018/3068365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chishti E.A., Edgington T.L., Chishti A.A., Ortiz-Soriano V., Neyra J.A. Severe acute kidney injury caused by decompression sickness syndrome. Clin Nephrol. 2022;97:298–304. doi: 10.5414/CN110662. [DOI] [PubMed] [Google Scholar]
- 23.Tang Z., Li T., Dai H., Feng C., Xie X., Peng F., et al. Drug-induced fanconi syndrome in patients with kidney allograft transplantation. Front Immunol. 2022;13 doi: 10.3389/fimmu.2022.979983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Chen J., Peng H., Chen C., Wang Y., Sang T., Cai Z., et al. Nag-1/gdf15 inhibits diabetic nephropathy via inhibiting age/rage-mediated inflammation signaling pathways in c57bl/6 mice and hk-2 cells. Life Sci. 2022;311 doi: 10.1016/j.lfs.2022.121142. [DOI] [PubMed] [Google Scholar]
- 25.Dong W., Zhang K., Gong Z., Luo T., Li J., Wang X., et al. N-acetylcysteine delayed cadmium-induced chronic kidney injury by activating the sirtuin 1-p53 signaling pathway. Chem Biol Inter. 2023;369 doi: 10.1016/j.cbi.2022.110299. [DOI] [PubMed] [Google Scholar]
- 26.Hou B., Liu M., Chen Y., Ni W., Suo X., Xu Y., et al. Cpd-42 protects against calcium oxalate nephrocalcinosis-induced renal injury and inflammation by targeting ripk3-mediated necroptosis. Front Pharm. 2022;13 doi: 10.3389/fphar.2022.1041117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Yan Q., Hu Q., Li G., Qiao Q., Ziyan S., Jie S., et al. Neat1 regulates caox crystal-induced renal tubular oxidative injury via mir-130/irf1. Antioxid Redox Signal. 2023;38:731–746. doi: 10.1089/ars.2022.0008. [DOI] [PubMed] [Google Scholar]
- 28.Zhou D., Wu Y., Yan H., Shen T., Li S., Gong J., et al. Gallic acid ameliorates calcium oxalate crystal-induced renal injury via upregulation of nrf2/ho-1 in the mouse model of stone formation. Phytomedicine. 2022;106 doi: 10.1016/j.phymed.2022.154429. [DOI] [PubMed] [Google Scholar]
- 29.Tang Y.W., Yang R.C., Wan F., Tang X.L., Zhang H.Q., Lin Y. Celastrol attenuates renal injury in 5/6 nephrectomized rats via inhibiting epithelial-mesenchymal transition and transforming growth factor-beta1/smad3 pathway. Exp Biol Med. 2022;247:1947–1955. doi: 10.1177/15353702221118087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Pei M.X., Dong S.J., Gao X.Y., Luo T., Fan D., Jin J.F., et al. Salvianolic acid b attenuates iopromide-induced renal tubular epithelial cell injury by inhibiting the tlr4/nf-kappab/nlrp3 signaling pathway. Evid Based Complement Altern Med. 2022;2022 doi: 10.1155/2022/8400496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Chaiyarit S., Thongboonkerd V. Oxidized forms of uromodulin promote calcium oxalate crystallization and growth, but not aggregation. Int J Biol Macromol. 2022;214:542–553. doi: 10.1016/j.ijbiomac.2022.06.132. [DOI] [PubMed] [Google Scholar]
- 32.Chaiyarit S., Thongboonkerd V. Oxidative modifications switch modulatory activities of urinary proteins from inhibiting to promoting calcium oxalate crystallization, growth, and aggregation. Mol Cell Proteom. 2021;20 doi: 10.1016/j.mcpro.2021.100151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Yoodee S., Noonin C., Sueksakit K., Kanlaya R., Chaiyarit S., Peerapen P., et al. Effects of secretome derived from macrophages exposed to calcium oxalate crystals on renal fibroblast activation. Commun Biol. 2021;4 doi: 10.1038/s42003-021-02479-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wuttimongkolchai N., Kanlaya R., Nanthawuttiphan S., Subkod C., Thongboonkerd V. Chlorogenic acid enhances endothelial barrier function and promotes endothelial tube formation: a proteomics approach and functional validation. Biomed Pharm. 2022;153 doi: 10.1016/j.biopha.2022.113471. [DOI] [PubMed] [Google Scholar]
- 35.Tyanova S., Temu T., Cox J. The maxquant computational platform for mass spectrometry-based shotgun proteomics. Nat Protoc. 2016;11:2301–2319. doi: 10.1038/nprot.2016.136. [DOI] [PubMed] [Google Scholar]
- 36.Cox J., Hein M.Y., Luber C.A., Paron I., Nagaraj N., Mann M. Accurate proteome-wide label-free quantification by delayed normalization and maximal peptide ratio extraction, termed maxlfq. Mol Cell Proteom. 2014;13:2513–2526. doi: 10.1074/mcp.M113.031591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Thevenot E.A., Roux A., Xu Y., Ezan E., Junot C. Analysis of the human adult urinary metabolome variations with age, body mass index, and gender by implementing a comprehensive workflow for univariate and opls statistical analyses. J Proteome Res. 2015;14:3322–3335. doi: 10.1021/acs.jproteome.5b00354. [DOI] [PubMed] [Google Scholar]
- 38.Sintiprungrat K., Singhto N., Thongboonkerd V. Characterization of calcium oxalate crystal-induced changes in the secretome of u937 human monocytes. Mol Biosyst. 2016;12:879–889. doi: 10.1039/c5mb00728c. [DOI] [PubMed] [Google Scholar]
- 39.Thanomkitti K., Kanlaya R., Fong-ngern K., Kapincharanon C., Sueksakit K., Chanchaem P., et al. Differential proteomics of lesional vs. Non-lesional biopsies revealed non-immune mechanisms of alopecia areata. Sci Rep. 2018;8 doi: 10.1038/s41598-017-18282-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Thongboonkerd V., Semangoen T., Chutipongtanate S. Factors determining types and morphologies of calcium oxalate crystals: molar concentrations, buffering, ph, stirring and temperature. Clin Chim Acta. 2006;367:120–131. doi: 10.1016/j.cca.2005.11.033. [DOI] [PubMed] [Google Scholar]
- 41.Thongboonkerd V., Chutipongtanate S., Semangoen T., Malasit P. Urinary trefoil factor 1 is a novel potent inhibitor of calcium oxalate crystal growth and aggregation. J Urol. 2008;179:1615–1619. doi: 10.1016/j.juro.2007.11.041. [DOI] [PubMed] [Google Scholar]
- 42.Aluksanasuwan S., Plumworasawat S., Malaitad T., Chaiyarit S., Thongboonkerd V. High glucose induces phosphorylation and oxidation of mitochondrial proteins in renal tubular cells: A proteomics approach. Sci Rep. 2020;10 doi: 10.1038/s41598-020-62665-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Nilnumkhum A., Kanlaya R., Yoodee S., Thongboonkerd V. Caffeine inhibits hypoxia-induced renal fibroblast activation by antioxidant mechanism. Cell Adh Migr. 2019;13:260–272. doi: 10.1080/19336918.2019.1638691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kanlaya R., Kapincharanon C., Fong-Ngern K., Thongboonkerd V. Induction of mesenchymal-epithelial transition (met) by epigallocatechin-3-gallate to reverse epithelial-mesenchymal transition (emt) in snai1-overexpressed renal cells: a potential anti-fibrotic strategy. J Nutr Biochem. 2022;107 doi: 10.1016/j.jnutbio.2022.109066. [DOI] [PubMed] [Google Scholar]
- 45.Fong-ngern K., Ausakunpipat N., Singhto N., Sueksakit K., Thongboonkerd V. Prolonged k(+) deficiency increases intracellular atp, cell cycle arrest and cell death in renal tubular cells. Metabolism. 2017;74:47–61. doi: 10.1016/j.metabol.2016.12.014. [DOI] [PubMed] [Google Scholar]
- 46.Peerapen P., Thongboonkerd V. Calcium oxalate monohydrate crystal disrupts tight junction via f-actin reorganization. Chem Biol Inter. 2021;345 doi: 10.1016/j.cbi.2021.109557. [DOI] [PubMed] [Google Scholar]
- 47.Hadpech S., Peerapen P., Thongboonkerd V. Alpha-tubulin relocalization is involved in calcium oxalate-induced tight junction disruption in renal epithelial cells. Chem Biol Inter. 2022;368 doi: 10.1016/j.cbi.2022.110236. [DOI] [PubMed] [Google Scholar]
- 48.Pongsakul N., Vinaiphat A., Chanchaem P., Fong-ngern K., Thongboonkerd V. Lamin a/c in renal tubular cells is important for tissue repair, cell proliferation, and calcium oxalate crystal adhesion, and is associated with potential crystal receptors. FASEB J. 2016;30:3368–3377. doi: 10.1096/fj.201600426R. [DOI] [PubMed] [Google Scholar]
- 49.Vinaiphat A., Thongboonkerd V. Characterizations of pmca2-interacting complex and its role as a calcium oxalate crystal-binding protein. Cell Mol Life Sci. 2018;75:1461–1482. doi: 10.1007/s00018-017-2699-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kanlaya R., Peerapen P., Nilnumkhum A., Plumworasawat S., Sueksakit K., Thongboonkerd V. Epigallocatechin-3-gallate prevents tgf-beta1-induced epithelial-mesenchymal transition and fibrotic changes of renal cells via gsk-3beta/beta-catenin/snail1 and nrf2 pathways. J Nutr Biochem. 2020;76 doi: 10.1016/j.jnutbio.2019.108266. [DOI] [PubMed] [Google Scholar]
- 51.Thanomkitti K., Fong-ngern K., Sueksakit K., Thuangtong R., Thongboonkerd V. Molecular functional analyses revealed essential roles of hsp90 and lamin a/c in growth, migration, and self-aggregation of dermal papilla cells. Cell Death Discov. 2018;4 doi: 10.1038/s41420-018-0053-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Midttun O., Ulvik A., Nygard O., Ueland P.M. Performance of plasma trigonelline as a marker of coffee consumption in an epidemiologic setting. Am J Clin Nutr. 2018;107:941–947. doi: 10.1093/ajcn/nqy059. [DOI] [PubMed] [Google Scholar]
- 53.Bresciani L., Tassotti M., Rosi A., Martini D., Antonini M., Dei Cas A., et al. Absorption, pharmacokinetics, and urinary excretion of pyridines after consumption of coffee and cocoa-based products containing coffee in a repeated dose, crossover human intervention study. Mol Nutr Food Res. 2020;64 doi: 10.1002/mnfr.202000489. [DOI] [PubMed] [Google Scholar]
- 54.Tanveer M.A., Rashid H., Nazir L.A., Archoo S., Shahid N.H., Ragni G., et al. Trigonelline, a plant derived alkaloid prevents ultraviolet-b-induced oxidative DNA damage in primary human dermal fibroblasts and balb/c mice via modulation of phosphoinositide 3-kinase-akt-nrf2 signalling axis. Exp Gerontol. 2023;171 doi: 10.1016/j.exger.2022.112028. [DOI] [PubMed] [Google Scholar]
- 55.Chen C., Shi Y., Ma J., Chen Z., Zhang M., Zhao Y. Trigonelline reverses high glucose-induced proliferation, fibrosis of mesangial cells via modulation of wnt signaling pathway. Diabetol Metab Syndr. 2022;14:28. doi: 10.1186/s13098-022-00798-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Zhang W., Zhang Y., Chen S., Zhang H., Yuan M., Xiao L., et al. Trigonelline, an alkaloid from Leonurus japonicus houtt., suppresses mast cell activation and ova-induced allergic asthma. Front Pharm. 2021;12 doi: 10.3389/fphar.2021.687970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Fouzder C., Mukhuty A., Mukherjee S., Malick C., Kundu R. Trigonelline inhibits nrf2 via egfr signalling pathway and augments efficacy of cisplatin and etoposide in nsclc cells. Toxicol Vitr. 2021;70 doi: 10.1016/j.tiv.2020.105038. [DOI] [PubMed] [Google Scholar]
- 58.Peerapen P., Chaiyarit S., Thongboonkerd V. Protein network analysis and functional studies of calcium oxalate crystal-induced cytotoxicity in renal tubular epithelial cells. Proteomics. 2018;18 doi: 10.1002/pmic.201800008. [DOI] [PubMed] [Google Scholar]
- 59.Sun X.Y., Gan Q.Z., Ouyang J.M. Calcium oxalate toxicity in renal epithelial cells: the mediation of crystal size on cell death mode. Cell Death Discov. 2015;1 doi: 10.1038/cddiscovery.2015.55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kogure K., Manabe S., Suzuki I., Tokumura A., Fukuzawa K. Cytotoxicity of alpha-tocopheryl succinate, malonate and oxalate in normal and cancer cells in vitro and their anti-cancer effects on mouse melanoma in vivo. J Nutr Sci Vitam. 2005;51:392–397. doi: 10.3177/jnsv.51.392. [DOI] [PubMed] [Google Scholar]
- 61.Chen C., Ma J., Miao C.S., Zhang H., Zhang M., Cao X., et al. Trigonelline induces autophagy to protect mesangial cells in response to high glucose via activating the mir-5189-5p-ampk pathway. Phytomedicine. 2021;92 doi: 10.1016/j.phymed.2021.153614. [DOI] [PubMed] [Google Scholar]
- 62.Sharma L., Lone N.A., Knott R.M., Hassan A., Abdullah T. Trigonelline prevents high cholesterol and high fat diet induced hepatic lipid accumulation and lipo-toxicity in c57bl/6j mice, via restoration of hepatic autophagy. Food Chem Toxicol. 2018;121:283–296. doi: 10.1016/j.fct.2018.09.011. [DOI] [PubMed] [Google Scholar]
- 63.Ilavenil S., Kim D.H., Jeong Y.I., Arasu M.V., Vijayakumar M., Prabhu P.N., et al. Trigonelline protects the cardiocyte from hydrogen peroxide induced apoptosis in h9c2 cells. Asian Pac J Trop Med. 2015;8:263–268. doi: 10.1016/S1995-7645(14)60328-X. [DOI] [PubMed] [Google Scholar]
- 64.Worley B., Powers R. Multivariate analysis in metabolomics. Curr Metab. 2013;1:92–107. doi: 10.2174/2213235X11301010092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Watanabe Y., Hirao Y., Kasuga K., Kitamura K., Nakamura K., Yamamoto T. Urinary proteome profiles associated with cognitive decline in community elderly residents-a pilot study. Front Neurol. 2023;14 doi: 10.3389/fneur.2023.1134976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Tang J., Wang Y., Luo Y., Fu J., Zhang Y., Li Y., et al. Computational advances of tumor marker selection and sample classification in cancer proteomics. Comput Struct Biotechnol J. 2020;18:2012–2025. doi: 10.1016/j.csbj.2020.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Chang Q., Chen P., Yin J., Liang G., Dai Y., Guan Y., et al. Discovery and validation of bladder cancer related excreted nucleosides biomarkers by dilution approach in cell culture supernatant and urine using uhplc-ms/ms. J Proteom. 2023;270 doi: 10.1016/j.jprot.2022.104737. [DOI] [PubMed] [Google Scholar]
- 68.Xing L., Sun L., Liu S., Zhang L., Sun J., Yang H. Metabolomic analysis of white, green and purple morphs of sea cucumber Apostichopus japonicus during body color pigmentation process. Comp Biochem Physiol Part D Genom Proteom. 2021;39 doi: 10.1016/j.cbd.2021.100827. [DOI] [PubMed] [Google Scholar]
- 69.Cheung H.W., Wong K.S., To N.S., Bond A.J., Farrington A.F., Prabhu A., et al. Label-free proteomics for discovering biomarker candidates of rad140 administration to castrated horses. Drug Test Anal. 2021;13:1034–1047. doi: 10.1002/dta.2988. [DOI] [PubMed] [Google Scholar]
- 70.Chen Y., Ni J., Gao Y., Zhang J., Liu X., Chen Y., et al. Integrated proteomics and metabolomics reveals the comprehensive characterization of antitumor mechanism underlying shikonin on colon cancer patient-derived xenograft model. Sci Rep. 2020;10 doi: 10.1038/s41598-020-71116-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Ma H., Lai B., Jin Y., Tian C., Liu J., Wang K. Proteomics and metabolomics analysis reveal potential mechanism of extended-spectrum beta-lactamase production in Escherichia coli. RSC Adv. 2020;10:26862–26873. doi: 10.1039/d0ra04250a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Yang Q., Wang Q., Wang Z., Li Z., Tang H., Yan X., et al. Metabolomic profiling and antidiabetic activity of callerya speciosa. Nat Prod Res. 2023:1–5. doi: 10.1080/14786419.2023.2265535. [DOI] [PubMed] [Google Scholar]
- 73.Li Z.Q., Yin X.L., Gu H.W., Zou D., Ding B., Li Z., et al. Revealing the chemical differences and their application in the storage year prediction of qingzhuan tea by swath-ms based metabolomics analysis. Food Res Int. 2023;173 doi: 10.1016/j.foodres.2023.113238. [DOI] [PubMed] [Google Scholar]
- 74.Wang Y.L., Zhu M.Y., Yuan Z.F., Ren X.Y., Guo X.T., Hua Y., et al. Proteomic profiling of cerebrospinal fluid in pediatric myelin oligodendrocyte glycoprotein antibody-associated disease. World J Pedia. 2022 doi: 10.1007/s12519-022-00661-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kim H.J., Seong E.Y., Lee W., Kim S., Ahn H.S., Yeom J., et al. Comparative analysis of therapeutic effects between medium cut-off and high flux dialyzers using metabolomics and proteomics: exploratory, prospective study in hemodialysis. Sci Rep. 2021;11 doi: 10.1038/s41598-021-96974-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Thangaraj S., Palanisamy S.K., Zhang G., Sun J. Quantitative proteomic profiling of marine diatom skeletonema dohrnii in response to temperature and silicate induced environmental stress. Front Microbiol. 2020;11 doi: 10.3389/fmicb.2020.554832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Surma S., Sahebkar A., Banach M. Coffee or tea: anti-inflammatory properties in the context of atherosclerotic cardiovascular disease prevention. Pharm Res. 2022;187 doi: 10.1016/j.phrs.2022.106596. [DOI] [PubMed] [Google Scholar]
- 78.Elghouizi A., Al-Waili N., Elmenyiy N., Elfetri S., Aboulghazi A., Al-Waili A., et al. Protective effect of bee pollen in acute kidney injury, proteinuria, and crystalluria induced by ethylene glycol ingestion in rats. Sci Rep. 2022;12:8351. doi: 10.1038/s41598-022-12086-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Hoseinynejad K., Mard S.A., Mansouri Z., Lamoochi Z., Kazemzadeh R. Efficacy of chlorogenic acid against ethylene glycol-induced renal stone model: the role of nfkb-runx2-ap1-osterix signaling pathway. Tissue Cell. 2022;79 doi: 10.1016/j.tice.2022.101960. [DOI] [PubMed] [Google Scholar]
- 80.Wang J., Chen J.J., Huang J.H., Lv B.D., Huang X.J., Hu Q., et al. Protective effects of total flavonoids from lysimachia christinae on calcium oxalate-induced oxidative stress in a renal cell line and renal tissue. Evid Based Complement Altern Med. 2021;2021 doi: 10.1155/2021/6667902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Yuan P., Sun X., Liu X., Hutterer G., Pummer K., Hager B., et al. Kaempferol alleviates calcium oxalate crystal-induced renal injury and crystal deposition via regulation of the ar/nox2 signaling pathway. Phytomedicine. 2021;86 doi: 10.1016/j.phymed.2021.153555. [DOI] [PubMed] [Google Scholar]
- 82.Lone A.N., Malik A.T., Naikoo H.S., Raghu R.S. S AT. Trigonelline, a naturally occurring alkaloidal agent protects ultraviolet-b (uv-b) irradiation induced apoptotic cell death in human skin fibroblasts via attenuation of oxidative stress, restoration of cellular calcium homeostasis and prevention of endoplasmic reticulum (er) stress. J Photochem Photobio B. 2020;202 doi: 10.1016/j.jphotobiol.2019.111720. [DOI] [PubMed] [Google Scholar]
- 83.Lorigooini Z., Sadeghi Dehsahraei K., Bijad E., Habibian Dehkordi S., Amini-Khoei H. Trigonelline through the attenuation of oxidative stress exerts antidepressant- and anxiolytic-like effects in a mouse model of maternal separation stress. Pharmacology. 2020;105:289–299. doi: 10.1159/000503728. [DOI] [PubMed] [Google Scholar]
- 84.Costa M.C., Lima T.F.O., Arcaro C.A., Inacio M.D., Batista-Duharte A., Carlos I.Z., et al. Trigonelline and curcumin alone, but not in combination, counteract oxidative stress and inflammation and increase glycation product detoxification in the liver and kidney of mice with high-fat diet-induced obesity. J Nutr Biochem. 2020;76 doi: 10.1016/j.jnutbio.2019.108303. [DOI] [PubMed] [Google Scholar]
- 85.Omidi-Ardali H., Lorigooini Z., Soltani A., Balali-Dehkordi S., Amini-Khoei H. Inflammatory responses bridge comorbid cardiac disorder in experimental model of ibd induced by dss: protective effect of the trigonelline. Inflammopharmacology. 2019;27:1265–1273. doi: 10.1007/s10787-019-00581-w. [DOI] [PubMed] [Google Scholar]
- 86.Hao X., Bu W., Lv G., Xu L., Hou D., Wang J., et al. Disrupted mitochondrial homeostasis coupled with mitotic arrest generates antineoplastic oxidative stress. Oncogene. 2022;41:427–443. doi: 10.1038/s41388-021-02105-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Guo M., Zhang W., Niu S., Shang M., Chang X., Wu T., et al. Adaptive regulations of nrf2 alleviates silver nanoparticles-induced oxidative stress-related liver cells injury. Chem Biol Inter. 2023;369 doi: 10.1016/j.cbi.2022.110287. [DOI] [PubMed] [Google Scholar]
- 88.Nishio T., Kishi R., Sato K., Sato K. Blue light exposure enhances oxidative stress, causes DNA damage, and induces apoptosis signaling in b16f1 melanoma cells. Mutat Res Genet Toxicol Environ Mutagen. 2022;883–884 doi: 10.1016/j.mrgentox.2022.503562. [DOI] [PubMed] [Google Scholar]
- 89.Canaud G., Brooks C.R., Kishi S., Taguchi K., Nishimura K., Magassa S., et al. Cyclin g1 and tascc regulate kidney epithelial cell g(2)-m arrest and fibrotic maladaptive repair. Sci Transl Med. 2019;11 doi: 10.1126/scitranslmed.aav4754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Khamchun S., Thongboonkerd V. Cell cycle shift from g0/g1 to s and g2/m phases is responsible for increased adhesion of calcium oxalate crystals on repairing renal tubular cells at injured site. Cell Death Discov. 2018;4:106. doi: 10.1038/s41420-018-0123-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Hou J. The kidney tight junction (review) Int J Mol Med. 2014;34:1451–1457. doi: 10.3892/ijmm.2014.1955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Ji Y., Fang S., Yang Y., Wu Z. Inactivation of the wnt/beta-catenin signaling contributes to the epithelial barrier dysfunction induced by sodium oxalate in canine renal epithelial cells. J Anim Sci. 2021;99 doi: 10.1093/jas/skab268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Peerapen P., Thongboonkerd V. Effects of calcium oxalate monohydrate crystals on expression and function of tight junction of renal tubular epithelial cells. Lab Invest. 2011;91:97–105. doi: 10.1038/labinvest.2010.167. [DOI] [PubMed] [Google Scholar]
- 94.Peerapen P., Thongboonkerd V. P38 mapk mediates calcium oxalate crystal-induced tight junction disruption in distal renal tubular epithelial cells. Sci Rep. 2013;3 doi: 10.1038/srep01041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Casula E., Pisano M.B., Serreli G., Zodio S., Melis M.P., Corona G., et al. Probiotic lactobacilli attenuate oxysterols-induced alteration of intestinal epithelial cell monolayer permeability: focus on tight junction modulation. Food Chem Toxicol. 2022;172 doi: 10.1016/j.fct.2022.113558. [DOI] [PubMed] [Google Scholar]
- 96.Qin W., Xu B., Chen Y., Yang W., Xu Y., Huang J., et al. Dietary ellagic acid supplementation attenuates intestinal damage and oxidative stress by regulating gut microbiota in weanling piglets. Anim Nutr. 2022;11:322–333. doi: 10.1016/j.aninu.2022.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Lin J., Zuo C., Liang T., Huang Y., Kang P., Xiao K., et al. Lycopene alleviates multiple-mycotoxin-induced toxicity by inhibiting mitochondrial damage and ferroptosis in the mouse jejunum. Food Funct. 2022;13:11532–11542. doi: 10.1039/d2fo02994d. [DOI] [PubMed] [Google Scholar]
- 98.Dhondup T., Kittanamongkolchai W., Vaughan L.E., Mehta R.A., Chhina J.K., Enders F.T., et al. Risk of esrd and mortality in kidney and bladder stone formers. Am J Kidney Dis. 2018;72:790–797. doi: 10.1053/j.ajkd.2018.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Sheng L., Zhuang S. New insights into the role and mechanism of partial epithelial-mesenchymal transition in kidney fibrosis. Front Physiol. 2020;11 doi: 10.3389/fphys.2020.569322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Ning W., Xu X., Zhou S., Wu X., Wu H., Zhang Y., et al. Effect of high glucose supplementation on pulmonary fibrosis involving reactive oxygen species and tgf-beta. Front Nutr. 2022;9 doi: 10.3389/fnut.2022.998662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Kanlaya R., Subkod C., Nanthawuttiphan S., Thongboonkerd V. Caffeine prevents oxalate-induced epithelial-mesenchymal transition of renal tubular cells by its anti-oxidative property through activation of nrf2 signaling and suppression of snail1 transcription factor. Biomed Pharm. 2021;141 doi: 10.1016/j.biopha.2021.111870. [DOI] [PubMed] [Google Scholar]
- 102.He X., Li G., Chen Y., Xiao Q., Yu X., Yu X., et al. Pharmacokinetics and pharmacodynamics of the combination of rhein and curcumin in the treatment of chronic kidney disease in rats. Front Pharm. 2020;11 doi: 10.3389/fphar.2020.573118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ghule A.E., Jadhav S.S., Bodhankar S.L. Trigonelline ameliorates diabetic hypertensive nephropathy by suppression of oxidative stress in kidney and reduction in renal cell apoptosis and fibrosis in streptozotocin induced neonatal diabetic (nstz) rats. Int Immunopharmacol. 2012;14:740–748. doi: 10.1016/j.intimp.2012.10.004. [DOI] [PubMed] [Google Scholar]
- 104.Peerapen P., Thongboonkerd V. Protective roles of trigonelline against oxalate-induced epithelial-to-mesenchymal transition in renal tubular epithelial cells: an in vitro study. Food Chem Toxicol. 2020;135 doi: 10.1016/j.fct.2019.110915. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplementary material








