Basic Study Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Aug 7, 2025; 31(29): 107066
Published online Aug 7, 2025. doi: 10.3748/wjg.v31.i29.107066
Tetrahydroxylated bile acids prevents malignant progression of Barret esophagus in vitro by inhibiting the interleukin-1β-nuclear factor kappa-B pathway
Anatolii Mamchur, Zhen-Wei Ma, Victor Ling, Zu-Hua Gao, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver V6R2B5, British Columbia, Canada
Shane Duggan, Department of Medicine, Rm 5.408 Life Sciences Institute, University of British Columbia, Vancouver V6T1Z3, British Columbia, Canada
Hui Xue, Xiao-Jia Niu, Department of Experimental Therapeutics, BC Cancer Research Center, Vancouver V5Z0B4, British Columbia, Canada
Yu-Zhuo Wang, Department of Urologic Sciences, Vancouver General Hospital, Vancouver V5Z0B4, British Columbia, Canada
Yu-Zhuo Wang, Department of Experimental Therapeutics, The University of British Columbia, Vancouver V5Z0B4, British Columbia, Canada
Dermot Kelleher, Department of Medicine, University of British Columbia, Vancouver V6T2B5, British Columbia, Canada
ORCID number: Zu-Hua Gao (0000-0001-7232-0933).
Co-first authors: Anatolii Mamchur and Shane Duggan.
Co-corresponding authors: Victor Ling and Zu-Hua Gao.
Author contributions: Mamchur A contributed to design and performance of experiments, data collection and analysis, manuscript writing, interpretation of data; Duggan S contributed to design of experiments, revision and editing of manuscript; Xue H and Niu XJ contributed to revision and editing of manuscript, methodology; Wang YZ and Ma ZW contributed to analysis, interpretation of data, revision and editing of manuscript; Kelleher D contributed to data analysis, revision and editing of manuscript, supervision of the study; Ling V and Gao Z contributed to supervision of the study, design of experiments, manuscript writing, revision and editing of manuscript.
Institutional review board statement: This study does not involve any human experiments. The study has been approved by the institutional biosafety board (No. B22-0164).
Institutional animal care and use committee statement: No animal experiment.
Conflict-of-interest statement: The authors declare that they have no conflict of interest.
Data sharing statement: No additional data are available.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Zu-Hua Gao, MD, PhD, FRCPC, Professor and Head, Department of Pathology and Laboratory Medicine, University of British Columbia, G105-2211 Wesbrook Mall, Vancouver V6R2B5, British Columbia, Canada. zuhua.gao@ubc.ca
Received: March 18, 2025
Revised: April 29, 2025
Accepted: June 30, 2025
Published online: August 7, 2025
Processing time: 140 Days and 10.2 Hours

Abstract
BACKGROUND

Barrett esophagus (BE), a metaplastic adaptive process to gastrointestinal reflux, is associated with a higher risk of developing esophageal adenocarcinoma. However, the factors and mechanism that drive the malignant progression of BE is not well understood.

AIM

To investigate the role of bile acids, a component of the reflux fluid, in the malignant progression of BE.

METHODS

Using engineered green fluorescent protein- labeled adult tissue-resident stem cells isolated from BE clinical biopsies (BE-ASCs) as the target, we studied the effect of hydrophobic deoxycholic acid (DCA) and hydrophilic tetrahydroxylated bile acids (THBA) on cell viability by fluorescence intensity analysis, mucin production by dark density measurement, tissue structure by pathology analysis, expression of different pro-inflammatory factors gene by quantitative polymerase chain reaction and proteins by Western blot.

RESULTS

We found that hydrophobic DCA has cytotoxic and proinflammatory effects through activation of interleukin-1β (IL-1β)-nuclear factor kappa-B (NF-κB) inflammatory pathway on BE-ASCs. This action results in impaired cell viability, tissue intactness, reduced mucin production, and increased transition to disorganized atypical cells without intestinal features. In contrast, co-culture with hydrophilic THBA inhibited the IL-1β-NF-κB inflammatory pathway with maintenance of mature intestinal type cellular and histomorphology.

CONCLUSION

Our data indicates that the hydrophilic bile acid THBA can counteract the cytotoxic and proinflammatory effect of hydrophobic DCA and prevent the malignant progression of BE by inhibiting the IL-1β-NF-κB pathway.

Key Words: Barrett esophagus; Inflammation; Metaplasia; Mucin; Cell viability; Histology; Tetrahydroxylated bile acids; Deoxycholic bile acid; Pathway

Core Tip: Barrett’s esophagus is a pathological precancerous condition. Hydrophobic bile acids could stimulate processes of malignant transition of Barrett esophagus tissue via activation proinflammatory signaling pathways. This study demonstrated that hydrophilic tetrahydroxylated bile acid could inhibit the cytotoxic and proinflammatory action of hydrophobic deoxycholic acid in adult stem cells isolated from Barrett esophagus. Co-treatment with hydrophilic tetrahydroxylated bile acids maintained the metaplastic intestinal-type cells in their differentiated state with preserved histology and mucin production. Thus, tetrahydroxylated bile acids have a potential protective role against the malignant progression of Barrett esophagus.



INTRODUCTION

Gastroesophageal reflux disease (GERD) is caused by retrograde flow from the stomach into the esophagus. Refluxate contains an admixture of gastric and intestinal contents including a set of different bile acids. It is well known that long-term exposure to reflux leads to chronic inflammation of esophageal tissue and replacement of squamous esophageal epithelium with a mature intestine-like columnar epithelium, an adaptive process first described by Dr. Barrett[1] in 1950 and subsequent named “Barrett esophagus (BE)”. BE is considered a precancerous lesion and is closely related to the occurrence of esophageal adenocarcinoma[2-12]. However, it is still not clear what drives the progression from the adaptive metaplastic process to a malignant dysplastic process in the setting of BE.

Histologically dysplasia is characterized by loss of cell polarity, abnormal tissue architecture, and nuclear atypia[13]. Studies showed that gastric acid in the reflux content was not the primary cause of malignant progression of BE as inhibition of gastric acid secretion using proton pump inhibitors was unable to significantly reduce the incidence of esophageal carcinoma of BE patients[14-16]. On the other hand, hydrophobic bile acids were found to be one of the most important promoters of esophageal cancer initiation[17-21].

Recent studies showed that hydrophilic bile acid can counteract the effect of hydrophobic bile acid[17-20]. Previous investigation by our team and others showed that a hydrophilic bile acid, tetrahydroxylated bile acids (THBA), can mitigate liver damage and reduced formation of hepatocellular carcinoma in Mdr2-deficient mice that has high levels of toxic bile acids[22,23]. However, the potential role of THBA in the setting of BE has not been reported. Besides, THBA are the most potent hydrophilic bile acids characterized in mammals. THBA are significantly more hydrophilic comparatively to ursodeoxycholic acid (UDCA)[24], which is one of the most investigated hydrophilic bile acids and approved for clinical use by the United States Food and Drug Administration[25]. Thus, there was an increased interest in investigating THBA efficiency in contexts of increased hydrophilicity of this bile acid towards the clinically approved UDCA. In the study, we investigated whether the hydrophilic THBA can counteract the toxic hydrophobic deoxycholic bile acid in the gastroesophageal refluxate and protect the esophageal tissue from dysplasia, cancer initiation and progression.

MATERIALS AND METHODS
Cells culture and cytotoxicity study

This study was conducted using engineered green fluorescent protein-labeled (GFP) adult tissue-resident stem cells isolated from BE clinical biopsies (BE-ASCs) following approval by the institutional research ethics board (No. B22-0164). The study abides to the principles of the declaration of Helsinki. BE-ASCs were isolated from the clinical biopsies tissue using a modified green protocol, by digestion in dispase, collagenase type I, trypsin, and DNAse with ROCK inhibitor Y-27632 (Selleckchem) at 37 °C for 1 hour with agitation[26-29]. Cells were cultivated on Matrigel (Corning) and irradiated mouse embryonic 3T3 fibroblasts (Kerafast) feeder layer in optimized expansion culture Chinese Family Assessment Device media contained: 2.5 mmol/L Rock-inhibitor (Calbiochem, United States), 10 mmol/L nicotinamide (Sigma-Aldrich, United States), 1 mmol/L Jagged-1 (AnaSpec Inc, United States), 125 ng/mLR-spondin1 (RD systems, United States), 100 ng/mL human Noggin (Peprotech, United States) and 2 mmol/L SB431542 (Cayman Chemical, United States). Bile acids cytotoxicity was assessed in three dimensional conditions (in 6.5 mm, pore size of 0.4 μm transwell; Corning) according to an optimized experimental scheme (Figure 1). 1 × 106 BE-ASCs were seeded into 12-transwells coated with 20% v/v Matrigel in cold phosphate-buffered saline and irradiated 3T3 mouse embryonic fibroblasts (1 × 105 fibroblasts per transwell). Briefly, BE-ASCs were cultured in an expansion cell medium for 4 days, then induced by air liquid interface (ALI) for 1 day and cultured in differentiation medium contained the same compounds excluding nicotinamide (Figure 1). Under these conditions BE-ASCs can form structures recapitulating Barrettes esophagus tissue (fragments of columnar well-polarized cells and interspersed goblet-like cells).

Figure 1
Figure 1 Scheme of cultivation of adult tissue-resident stem cells isolated from Barrett esophagus clinical biopsies and cytotoxicity investigation using different concentrations of deoxycholic acid, tetrahydroxylated bile acids and their combinations. BE-ASCs: Barrett esophagus clinical biopsies; DCA: Deoxycholic acid; THBA: Tetrahydroxylated bile acids.

On day 6, the cells were treated with different concentration of DCA in differentiating medium (10, 20, 30, 40, 50, 100, 150, 200, 400 μM) and synthetic THBA (200-5000 μM) to determine the optimal concentrations for treatment. Viability was measured after 2 weeks of cultivation. To evaluate viability, the levels of GFP fluorescence were measured and analyzed by using ImageJ software.

Mucin production

BE-ASCs were cultured in the same manner as described above. Levels of mucin production were measured on day 15 of BE-ASCs cultivation either without addition of bile acids (control) or treated with DCA, THBA and their combinations. Measurement was performed using ImageJ software.

Histological analysis

Inserts of membranes with BE-ASC ALI tissues were taken for fixation in 4% paraformaldehyde (15 minutes). All procedures [dehydration, paraffin-embedding, sectioning of ALI tissue, rehydration and hematoxylin and eosin (HE) staining of ALI tissue sections] were performed according to the protocol of Manna and Caradonna[30].

Proinflammatory gene analysis

Quantitative polymerase chain reaction (qPCR) was used to study the expression of proinflammatory genes [interleukin (IL)-6, IL-1β, tumor necrosis factor (TNF)-1α, cyclooxygenase-2 (COX-2)], and MUC-2 (intestinal marker) in BE-ASCs under the influence of DCA, THBA and their combination. BE-ASCs were cultured using optimized media with or w/o 100 μM DCA, 2000 μM THBA and their combination (2000 μM THBA/100 μM DCA). Total cellular RNA was isolated after 48 hours cultivation treatment using RNeasy mini kit (QIAGEN) according to the manufacturer’s protocol. The RNA was quantified on the Nano Drop 1000 where the quality of message RNA was evaluated by the optical density (OD)260/OD280 ratio. For reverse transcription reaction, 1 μg of total RNA (high capacity completely DNA Archive kit) was used and 10 ng of transcribed DNA was spent for each qPCR reaction. As target probes, TaqMan IL-6, IL-1β, TNF-1α, COX-2 were used along with a housekeeping gene glyceraldehyde-3-phosphate dehydrogenase (GAPDH).

Proinflammatory protein analysis

Detection of cleaved (active form) of IL-1β and nuclear factor kappa-B (NF-κB) was conducted using Western blotting. Briefly, BE-ASCs were cultivated with optimized media with or w/o DCA 100 μM, THBA 2000 μM and their combination. Cellular lysates were obtained after 48 hours of cultivation using radioimmunoprecipitation assay lysis buffer (Millipore Sigma) and HaltTM Protease Phosphatase Inhibitor Cocktail (Thermo Scientific) according to the manufacturer’s protocol. The total protein concentration in the BE-ASCs lysates was detected in a 96-well plate using a Pierce™ bicinchoninic acid protein assay kit (Thermo Scientific) according to the manufacturer’s protocol. Western blot was performed using the protocol of Mahmood and Yang[31]. The following rabbit primary antibodies were used: IL-1β, NF-κB, GAPDH, β-actin and horseradish peroxidase-labeled goat anti-rabbit secondary antibody (Cell Signaling). Incubation membranes with antibodies were conducted according to the manufacturer’s protocol.

Statistical analysis

The results were represented as mean ± SD, and calculated with a two-tailed Student’s t test. Statistical significance was defined as P value of < 0.05, < 0.01, < 0.001 and < 0.0001. All experiments were performed using at least three biological replicates.

RESULTS
THBA protects the viability of DCA-treated BE-ASCs cells

To determine if THBA can protect BE-ASCs from the cellular damage caused by DCA, we tested the dose-dependent effect of each bile acid on these cells (Figure 2). As can be seen in Figure 2A and D, there was no decrease in viability in BE-ASCs at concentrations of up to 50 μM of DCA but there was visible cytotoxicity starting at 100 μM and a significant loss of viability (22-fold) when cells were treated with 150 μM or higher. Since the goal was to avoid extreme toxicity and possible non-specific effects, we chose to use 100 μM as the effective concentration of DCA to be used for this study.

Figure 2
Figure 2 Effect of deoxycholic acid, tetrahydroxylated bile acids and their combination on adult tissue-resident stem cells isolated from Barrett esophagus clinical biopsies cytotoxicity. Representative green fluorescent protein-labeled (GFP) fluorescent images of Barrett esophagus clinical biopsies (BE-ASCs) in transwell inserts after 15 days in culture with various deoxycholic acid (DCA) and hydrophilic tetrahydroxylated bile acids (THBA) concentrations. 1 × 106 BE-ASCs were plated in 12 Matrigel coated transwell inserts on the feeder layer of irradiated 3T3 mouse embryonic fibroblasts (1 × 105 fibroblasts per transwell). A: BE-ASCs cultured with 100 μM DCA showed slight cytotoxicity and significant decreasing of viability (22-fold) after treatment with 150 μM DCA or higher; B: BE-ASCs treated with 3000 μM THBA or higher demonstrated significant decreases in viability; C: BE-ASCs co-treated with 2000 μM THBA/150 μM DCA showed 17-fold higher viability in comparison to cells treated with DCA alone; D-F: Viability levels of BE-ASCs treated with DCA (D), THBA (E), their combination (F), were correlated with GFP fluorescence intensity measured using ImageJ software. Averages from triplicates are presented as mean ± SD (n = 3). cP < 0.001. dP < 0.0001. DCA: Deoxycholic acid; THBA: Tetrahydroxylated bile acids.

In a similar manner, we determined the highest non-toxic concentration of THBA that can be used in this system. As can be seen in Figure 2B and E no significant cytotoxicity was observed in BE-ASCs treated with THBA using concentrations ranging from 200-2000 μM; however, significant cytotoxicity was observed at 3000 μM and increasing to greater than 20 fold at 4000 and 5000 μM of THBA. We therefore chose 2000 μM of THBA to test the potential protective effect of this bile acid.

We next tested if the presence of THBA can protect EB-ASCs from the cellular damage caused by DCA using these two elected concentrations of bile acids (Figure 2C and F). We observed that co-treatment with 2000 μM THBA/150 μM DCA resulted in significantly higher cell viability (17-fold) compared with treatment with DCA alone.

THBA counteracted the inhibitory effect of DCA on BE-ASCs mucin production

Mucin production is an important gastro-intestinal functional indicator of the mucosa tissue and plays an important role in tissue protection. We investigated whether THBA could preserve mucin secretion in BE-ASCs exposed to DCA. As demonstrated (Figure 3), 150 μM DCA inhibits mucin secretion in BE-ASCs. The same cells co-treated with THBA (2000 μM) restored mucin production (the level of secretion was 17-fold higher in comparison to the cells treated with 150 μM DCA alone).

Figure 3
Figure 3 Effects of deoxycholic acid and tetrahydroxylated bile acids co-exposure on adult tissue-resident stem cells isolated from Barrett esophagus clinical biopsies mucin production. Representative bright field images of Barrett esophagus clinical biopsies (BE-ASCs) in transwell inserts. A: BE-ASCs treated with 2000 μM tetrahydroxylated bile acids restored mucin production (the level of secretion was 17-fold higher in comparison to the cells treated with 150 μM deoxycholic acid alone); B: Mucin levels were correlated with dark density measured by ImageJ software. Data are presented as mean ± SD (n = 3). dP < 0.0001. DCA: Deoxycholic acid; THBA: Tetrahydroxylated bile acids.
THBA supports intestinal differentiation in DCA-treated BE-ASCs

Inflammation, cellular disarray, tissue fragmentation and loss of goblet cells are early histological signs of progression from metaplasia to dysplasia. We investigated the effects of DCA and THBA on tissue histology using a model of ALI with transwell inserts. HE-stained tissue sections revealed a high level of differentiation in samples treated with THBA and control. There were interspersed goblet-like cells between well polarized columnar intestine-like cells resembling mature intestinal mucosa. Samples treated with DCA showed attenuation of the cell layer and reduced number of goblet-like cells, which becomes more severe with increasing DCA concentration. BE-ASCs treated with combination DCA/THBA showed high levels of intestinal differentiation. (Figure 4). Further, we examined the expression of MUC2, a molecular marker of intestinal differentiation. As shown in Figure 5, DCA treatment decreased MUC2 expression by 2 to 2.3-fold compared to untreated BE-ASCs, while THBA co-treatment was able to significantly restore MUC2 expression in DCA treated cells to over 50 percent of control. These data indicate that THBA can counteract the damaging effect of DCA by preserving intestinal differentiation in the metaplastic mucosa.

Figure 4
Figure 4 Effects of deoxycholic acid and tetrahydroxylated bile acids co-exposure on adult tissue-resident stem cells isolated from Barrett esophagus clinical biopsies intestinal differentiation. Representative pictures of hematoxylin and eosin stained air liquid interface Barrett esophagus clinical biopsies tissue sections (× 20 magnification). Samples treated with 2000 μM tetrahydroxylated bile acids (THBA), co-treated with THBA and deoxycholic acid (DCA) (2000 μM THBA/150 μM DCA, 2000 μM THBA/100 μM DCA) and control demonstrated high level intestinal differentiation with distinct interspersed goblet-like cells between well polarized columnar intestine-like cells resembling mature intestinal mucosa. Samples treated with DCA alone showed attenuation of the cell layer, reduced number of goblet dells and nuclear atypia. (1goblet-like cells, 2columnar polarized intestine-like cells). DCA: Deoxycholic acid; THBA: Tetrahydroxylated bile acids; Scale bar = 250 μm.
Figure 5
Figure 5 Effects of deoxycholic acid and tetrahydroxylated bile acids co-exposure on MUC2 gene expression in adult tissue-resident stem cells isolated from Barrett esophagus clinical biopsies. Barrett esophagus clinical biopsies cells treated with deoxycholic acid (DCA) showed decreased level of MUC2 expression by 2.3-fold in comparison to control cells, while tetrahydroxylated bile acids co-treatment significantly restored MUC2 expression in DCA treated cells to over 50 percent of control. Data are presented as mean ± SD (n = 3). bP < 0.01. cP < 0.001. DCA: Deoxycholic acid; THBA: Tetrahydroxylated bile acids.
THBA inhibits IL-1β-NF-κB pathway in DCA-treated BE-ASCs

Since one of the most important actions of hydrophobic bile acids is inflammatory stimulation[32-35], we studied the influence of DCA on gene expression of the most important inflammatory markers (IL-1β, IL-6, COX-2, TNF-α) and effect of THBA in combined treatment of BE-ASCs. We did not observe significant differences in the gene expression IL-6, COX-2, TNF-α in the cells treated with DCA, THBA, their combination and control (Figure 6). However, we found significant 2-fold increase in IL-1β gene expression after 48 hours exposure with 100 μM DCA (Figure 6).

Figure 6
Figure 6 Effects of deoxycholic acid and tetrahydroxylated bile acids co-exposure on proinflammatory (interleukin-6, interleukin-1β, tumor necrosis factor-α, cyclooxygenase-2) genes expression in adult tissue-resident stem cells isolated from Barrett esophagus clinical biopsies. Barrett esophagus clinical biopsies treated with deoxycholic acid, tetrahydroxylated bile acids, their combination did not demonstrate significant difference in the gene expression interleukin-6, cyclooxygenase-2, and tumor necrosis factor-α. At the same time, these cells demonstrated significant 2-fold increase in interleukin-1β gene expression after cultivation with 100 μM deoxycholic acid. Data are presented as mean ± SD (n = 3). aP < 0.05. bP < 0.01. IL: Interleukin; TNF-α: Tumor necrosis factor-α; COX2: Cyclooxygenase-2; DCA: Deoxycholic acid; THBA: Tetrahydroxylated bile acids.

Western blot analysis demonstrated that IL-1β is expressed in its bioactive form (17 kDa) in BE-ASCs (Figure 7). Treatment with 100 μM DCA increases IL-1β protein expression in BE-ASCs by 2-fold versus non-treated cells. THBA plus DCA co-exposure resulted in significant decrease of IL-1β expression vs DCA alone (Figure 7A and B).

Figure 7
Figure 7 Effect of deoxycholic acid and tetrahydroxylated bile acids co-exposure on interleukin-1β-nuclear factor kappa-B pathway activity in adult tissue-resident stem cells isolated from Barrett esophagus clinical biopsies. Representative immunoblots demonstrating effects of deoxycholic acid (DCA) and tetrahydroxylated bile acids (THBA) co-exposure on interleukin-1β (IL-1β)-nuclear factor kappa-B (NF-κB) pathway activity in Barrett esophagus clinical biopsies (BE-ASCs). A: Cells treated with 100 μM DCA showed significant 2-fold increase of IL-1β protein expression in comparison to untreated cells. THBA and DCA co-exposure resulted in significant decrease of IL-1β expression vs DCA alone; B: Expression levels of IL-1β in BE-ASCs treated with 100 μM DCA, 2000 μM THBA and their combination-2000 μM THBA/100 μM DCA, were correlated with density levels measured by using ImageJ software; C: BE-ASCs treated with DCA demonstrated significant higher level of NF-κB phosphorylation in comparison to untreated cells and treated with combination THBA/DCA, THBA; D: Expression levels of NF-κB in BE-ASCs treated with 100 μM DCA, 2000 μM THBA and their combination-2000 μM THBA/100 μM DCA, were correlated with density levels measured by using ImageJ software. Data are presented as mean ± SD (n = 3). aP < 0.05. bP < 0.01. IL: Interleukin; DCA: Deoxycholic acid; THBA: Tetrahydroxylated bile acids; GAPDH: Glyceraldehyde-3-phosphate dehydrogenase; NF-κB: Nuclear factor kappa-B.

IL-1β inflammatory factor is one of the important participants of the NF-κB pathway which in turn is a central regulator of inflammation. NF-κB signaling promotes the development of different cancers including esophageal adenocarcinoma[36-39]. Using Western blot analysis, BE-ASCs exposed with DCA showed around 1.5 folds higher level of phosphorylated (active form p-p65) NF-κB comparatively to the cells treated with combination THBA/DCA, THBA and untreated cells (Figure 7C and D). These results suggest THBA may prevent dysplasia and esophageal cancer development through inhibiting the NF-κB signaling pathway.

DISCUSSION

BE is a process of trans differentiation from squamous cells into goblet-cell containing mature intestinal mucosa cells. The stem cells of the distal esophagus and gastro-esophageal junction are thought to be the origin of BE and may be affected by biliary reflux[40]. BE is considered an aberrant wound healing process that protects the damage from the reflux as the intestinal mucosa is supposedly more resistant to digestion and toxic effect of intestinal contents in the reflux[41]. However, the mechanisms that regulate wound healing have been shown to promote transformation and growth of malignant cells. As a matter of fact, chronic inflammation has been associated with malignant transformation in many tissues[42]. Timely blockage of the inflammatory process in the metaplastic mucosa has the potential to maintain tissue homeostasis and prevent the malignant progression in the setting of BE. In this study, using cell viability, mucin production, MUC2 expression, and histological analysis, we found that hydrophilic bile acid THBA could counteract the toxic effect of hydrophobic bile acid DCA on BE-ASCs. Further analysis using PCR and Western blot indicate that the protective effect of THBA is at least in part mediated through the inhibition of IL-1β-NF-κB inflammatory pathway. Our study provided experimental evidence that hydrophilic bile acid THBA has the potential to prevent malignant progression of BE through inhibition of the inflammatory process caused by continuous reflux and exposure to toxic hydrophobic bile acid in the reflux content.

Progression of BE to adenocarcinoma is strongly influenced by bile acid exposure[21]. The hydrophobic bile acid DCA, in particular, has been shown to be genotoxic to esophageal cells through inducing the release of reactive oxygen species within the cytoplasm of exposed cells[43-47]. In turn, DNA damage caused by DCA increase the possibility of oncogenic mutations. In addition, hydrophobic bile acids have the ability to induce damage in the cell membrane through lipid peroxidation, and induction of apoptosis through different pathways[45,48,49]. The mechanism of cytoprotective action of THBA needs further investigation. We assume that THBA can be involved in similar antioxidant mechanisms as another hydrophilic acid UDCA. It has been demonstrated that hepatocytes pretreated with UDCA increased level of thiol-containing proteins which effectively neutralized hydroxyl radicals[45,50].

The present investigation demonstrated the ability of THBA to commit BE-ASCs in intestinal state, with saving functional and morphological properties of intestinal (metaplastic) tissue. We have shown that THBA treatment restored in 17 times mucin production in DCA treated BE-ASCs, which was further confirmed at the molecular level using qPCR analysis of MUC2 gene expression. MUC2 expression was significantly higher in BE-ASCs treated with a combination of THBA/DCA compared to cells treated with DCA alone (Figure 5). Moreover, THBA treatment alone increased MUC2 gene expression compared to untreated BE-ASCs. These results are consistent with previous investigation demonstrating that DCA decreased mucin levels in differentiated enterocyte-like cells, which can be prevented by co-incubation with another hydrophilic tauroursodeoxycholic acid[44]. We speculate that stimulation of mucin production can prevent damage to the mucosal layer and protect underlying cells susceptible to toxins or carcinogens. Thus, THBA can attenuate the transition from metaplasia to dysplasia and cancer initiation induced by hydrophobic bile acid.

Further, the present investigation revealed that THBA could suppress the activity of IL-1β-NF-κB proinflammatory pathway induced by DCA. Similar phenomenon has been observed in previous investigations with using another hydrophilic acid UDCA which inhibited IL-1β and DCA induced activation of NF-κB in human colon cancer cells[49,51]. In addition, it has been shown that UDCA inhibits expression of the pro- inflammatory factors IL-1β and nitric oxide and suppressed NF-κB activation in lipopolysaccharides-exposed (inflammatory inducer) rat, a model of neuroinflammation associated with neurodegeneration[52,53]. Miura et al[54] revealed that UDCA can activate glucocorticoid receptor (GR) with consecutive suppression of NF-κB-dependent transcription through the intervention of interaction between GR and phosphorylated unit of NF-κB p65. This study suggests that THBA may exploit similar molecular mechanisms of inactivation of IL-1β-NF-κB pathways as UDCA.

One of the limitations of our study is its sample size. Although the experiment was repeated at least three times, further research with expanded sample size could help to improve the generalization of the results. Secondly, in vivo studies using gastroesophageal reflux-BE animal models are required to further validate these in vitro results. Thirdly, more in-depth molecular studies using advanced target discovery technologies such as proteomics and proteolysis-targeting chimeras probe can be applied to further explore the underlying specific molecular mechanism and targets of THBA[55]. Lastly, the safety and efficacy of THBA needs to be well studied in both small and large animals before introducing it into clinic trials. If proved safe and effective for preventing gastroesophageal cancer in the clinical setting, we will further explore the role of THBA in other types of cancer such as cancer of the gastric antrum or other diseases that affects the metabolism and secretion of bile acids.

CONCLUSION

In summary, we have demonstrated that the hydrophilic bile acid THBA could counteract the toxic effect of hydrophobic bile acid DCA, maintain BE tissue in metaplastic intestinal-like state and hamper transition to “oncogenic” phase: dysplasia and adenocarcinoma initiation. Our data indicate that the protective effect of THBA is at least in part mediated through the inhibition of IL-1β-NF-κB inflammatory pathway. If validated in preclinical trials, THBA could be a viable preventive regimen for patients with BE that has ongoing gastroesophageal reflux.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: Canada

Peer-review report's classification

Scientific Quality: Grade A, Grade B, Grade B, Grade B, Grade B

Novelty: Grade A, Grade B, Grade B, Grade B, Grade B

Creativity or Innovation: Grade A, Grade B, Grade B, Grade B, Grade B

Scientific Significance: Grade B, Grade B, Grade B, Grade B, Grade C

P-Reviewer: Qin SL; Ying GH; Zhang SW S-Editor: Fan M L-Editor: A P-Editor: Zhao YQ

References
1.  BARRETT NR. Chronic peptic ulcer of the oesophagus and 'oesophagitis'. Br J Surg. 1950;38:175-182.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 655]  [Cited by in RCA: 545]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
2.  Stawinski PM, Dziadkowiec KN, Kuo LA, Echavarria J, Saligram S. Barrett's Esophagus: An Updated Review. Diagnostics (Basel). 2023;13:321.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 12]  [Reference Citation Analysis (0)]
3.  Morales TG, Sampliner RE. Barrett's esophagus: update on screening, surveillance, and treatment. Arch Intern Med. 1999;159:1411-1416.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 41]  [Cited by in RCA: 33]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
4.  Chang JT, Katzka DA. Gastroesophageal reflux disease, Barrett esophagus, and esophageal adenocarcinoma. Arch Intern Med. 2004;164:1482-1488.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 44]  [Cited by in RCA: 47]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
5.  Guillem PG. How to make a Barrett esophagus: pathophysiology of columnar metaplasia of the esophagus. Dig Dis Sci. 2005;50:415-424.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 39]  [Cited by in RCA: 38]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
6.  Fléjou JF. Barrett's oesophagus: from metaplasia to dysplasia and cancer. Gut. 2005;54 Suppl 1:i6-12.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 133]  [Cited by in RCA: 123]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
7.  Zhang L, Sun B, Zhou X, Wei Q, Liang S, Luo G, Li T, Lü M. Barrett's Esophagus and Intestinal Metaplasia. Front Oncol. 2021;11:630837.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
8.  Vercauteren Drubbel A, Beck B. Dedifferentiation of esophageal progenitors in metaplasia and cancer. Mol Cell Oncol. 2021;8:1991758.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (0)]
9.  Haggitt RC. Barrett's esophagus, dysplasia, and adenocarcinoma. Hum Pathol. 1994;25:982-993.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 457]  [Cited by in RCA: 440]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
10.  Runge TM, Abrams JA, Shaheen NJ. Epidemiology of Barrett's Esophagus and Esophageal Adenocarcinoma. Gastroenterol Clin North Am. 2015;44:203-231.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 140]  [Cited by in RCA: 163]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
11.  Thiruvengadam SK, Tieu AH, Luber B, Wang H, Meltzer SJ. Risk Factors for Progression of Barrett's Esophagus to High Grade Dysplasia and Esophageal Adenocarcinoma. Sci Rep. 2020;10:4899.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 42]  [Cited by in RCA: 45]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
12.  Buttar NS, Wang KK, Sebo TJ, Riehle DM, Krishnadath KK, Lutzke LS, Anderson MA, Petterson TM, Burgart LJ. Extent of high-grade dysplasia in Barrett's esophagus correlates with risk of adenocarcinoma. Gastroenterology. 2001;120:1630-1639.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 313]  [Cited by in RCA: 264]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
13.  Giroux V, Rustgi AK. Metaplasia: tissue injury adaptation and a precursor to the dysplasia-cancer sequence. Nat Rev Cancer. 2017;17:594-604.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 163]  [Cited by in RCA: 228]  [Article Influence: 28.5]  [Reference Citation Analysis (0)]
14.  Quilty F, Byrne AM, Aird J, El Mashad S, Parra-Blanco A, Long A, Gilmer JF, Medina C. Impact of Deoxycholic Acid on Oesophageal Adenocarcinoma Invasion: Effect on Matrix Metalloproteinases. Int J Mol Sci. 2020;21:8042.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 2]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
15.  Sun D, Wang X, Gai Z, Song X, Jia X, Tian H. Bile acids but not acidic acids induce Barrett's esophagus. Int J Clin Exp Pathol. 2015;8:1384-1392.  [PubMed]  [DOI]
16.  Gillen P, Keeling P, Byrne PJ, Healy M, O'Moore RR, Hennessy TP. Implication of duodenogastric reflux in the pathogenesis of Barrett's oesophagus. Br J Surg. 1988;75:540-543.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 127]  [Cited by in RCA: 126]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
17.  Peng S, Huo X, Rezaei D, Zhang Q, Zhang X, Yu C, Asanuma K, Cheng E, Pham TH, Wang DH, Chen M, Souza RF, Spechler SJ. In Barrett's esophagus patients and Barrett's cell lines, ursodeoxycholic acid increases antioxidant expression and prevents DNA damage by bile acids. Am J Physiol Gastrointest Liver Physiol. 2014;307:G129-G139.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 44]  [Cited by in RCA: 49]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
18.  Hu Y, Ye X, Wang R, Poon K. Current research progress in the role of reactive oxygen species in esophageal adenocarcinoma. Transl Cancer Res. 2021;10:1568-1577.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
19.  Hanafi NI, Mohamed AS, Sheikh Abdul Kadir SH, Othman MHD. Overview of Bile Acids Signaling and Perspective on the Signal of Ursodeoxycholic Acid, the Most Hydrophilic Bile Acid, in the Heart. Biomolecules. 2018;8:159.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 64]  [Cited by in RCA: 85]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
20.  Banerjee B, Shaheen NJ, Martinez JA, Hsu CH, Trowers E, Gibson BA, Della'Zanna G, Richmond E, Chow HH. Clinical Study of Ursodeoxycholic Acid in Barrett's Esophagus Patients. Cancer Prev Res (Phila). 2016;9:528-533.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 16]  [Cited by in RCA: 14]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
21.  Bernstein H, Bernstein C, Payne CM, Dvorak K. Bile acids as endogenous etiologic agents in gastrointestinal cancer. World J Gastroenterol. 2009;15:3329-3340.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 196]  [Cited by in RCA: 230]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
22.  Wang R, Sheps JA, Liu L, Han J, Chen PSK, Lamontagne J, Wilson PD, Welch I, Borchers CH, Ling V. Hydrophilic bile acids prevent liver damage caused by lack of biliary phospholipid in Mdr2(-/-) mice. J Lipid Res. 2019;60:85-97.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 26]  [Cited by in RCA: 29]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
23.  Sheps JA, Wang R, Wang J, Ling V. The protective role of hydrophilic tetrahydroxylated bile acids (THBA). Biochim Biophys Acta Mol Cell Biol Lipids. 2021;1866:158925.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
24.  Liu T, Wang RX, Han J, Li ZD, Sheps JA, Zheng LJ, Xu XX, Ling V, Wang JS. Comprehensive Bile Acid Profiling of ABCB4-mutated Patients and the Prognostic Role of Taurine-conjugated 3α,6α,7α,12α-Tetrahydroxylated Bile Acid in Cholestasis. J Clin Transl Hepatol. 2024;12:151-161.  [PubMed]  [DOI]  [Full Text]
25.  Rudic JS, Poropat G, Krstic MN, Bjelakovic G, Gluud C. Ursodeoxycholic acid for primary biliary cirrhosis. Cochrane Database Syst Rev. 2012;12:CD000551.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 34]  [Cited by in RCA: 54]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
26.  Duleba M, Yamamoto Y, Neupane R, Rao W, Xie J, Qi Y, Liew AA, Niroula S, Zhang Y, Mahalingam R, Wang S, Goller K, Ajani JA, Vincent M, Ho KY, Hou JK, Hyams JS, Sylvester FA, Crum CP, McKeon F, Xian W. Cloning of ground-state intestinal stem cells from endoscopic biopsy samples. Nat Protoc. 2020;15:1612-1627.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 3]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
27.  Wang X, Yamamoto Y, Wilson LH, Zhang T, Howitt BE, Farrow MA, Kern F, Ning G, Hong Y, Khor CC, Chevalier B, Bertrand D, Wu L, Nagarajan N, Sylvester FA, Hyams JS, Devers T, Bronson R, Lacy DB, Ho KY, Crum CP, McKeon F, Xian W. Cloning and variation of ground state intestinal stem cells. Nature. 2015;522:173-178.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 126]  [Cited by in RCA: 149]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
28.  Duleba M, Qi Y, Mahalingam R, Liew AA, Neupane R, Flynn K, Rinaldi F, Vincent M, Crum CP, Ho KY, Hou JK, Hyams JS, Sylvester FA, McKeon F, Xian W. An Efficient Method for Cloning Gastrointestinal Stem Cells From Patients via Endoscopic Biopsies. Gastroenterology. 2019;156:20-23.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 4]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
29.  Green H. The birth of therapy with cultured cells. Bioessays. 2008;30:897-903.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 115]  [Cited by in RCA: 105]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
30.  Manna V, Caradonna S. Isolation, expansion, differentiation, and histological processing of human nasal epithelial cells. STAR Protoc. 2021;2:100782.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
31.  Mahmood T, Yang PC. Western blot: technique, theory, and trouble shooting. N Am J Med Sci. 2012;4:429-434.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 491]  [Cited by in RCA: 759]  [Article Influence: 58.4]  [Reference Citation Analysis (0)]
32.  Quilty F, Freeley M, Gargan S, Gilmer J, Long A. Deoxycholic acid induces proinflammatory cytokine production by model oesophageal cells via lipid rafts. J Steroid Biochem Mol Biol. 2021;214:105987.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
33.  Phelan JP, Reen FJ, Caparros-Martin JA, O'Connor R, O'Gara F. Rethinking the bile acid/gut microbiome axis in cancer. Oncotarget. 2017;8:115736-115747.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 23]  [Cited by in RCA: 36]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
34.  Jang JY, Im E, Choi YH, Kim ND. Mechanism of Bile Acid-Induced Programmed Cell Death and Drug Discovery against Cancer: A Review. Int J Mol Sci. 2022;23:7184.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 12]  [Cited by in RCA: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
35.  Wei S, Ma X, Zhao Y. Mechanism of Hydrophobic Bile Acid-Induced Hepatocyte Injury and Drug Discovery. Front Pharmacol. 2020;11:1084.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 10]  [Cited by in RCA: 41]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
36.  Abdel-Latif MM, O'Riordan J, Windle HJ, Carton E, Ravi N, Kelleher D, Reynolds JV. NF-kappaB activation in esophageal adenocarcinoma: relationship to Barrett's metaplasia, survival, and response to neoadjuvant chemoradiotherapy. Ann Surg. 2004;239:491-500.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 159]  [Cited by in RCA: 168]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
37.  Li B, Li YY, Tsao SW, Cheung AL. Targeting NF-kappaB signaling pathway suppresses tumor growth, angiogenesis, and metastasis of human esophageal cancer. Mol Cancer Ther. 2009;8:2635-2644.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 74]  [Cited by in RCA: 87]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
38.  Xia Y, Shen S, Verma IM. NF-κB, an active player in human cancers. Cancer Immunol Res. 2014;2:823-830.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 658]  [Cited by in RCA: 909]  [Article Influence: 90.9]  [Reference Citation Analysis (0)]
39.  Taniguchi K, Karin M. NF-κB, inflammation, immunity and cancer: coming of age. Nat Rev Immunol. 2018;18:309-324.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1132]  [Cited by in RCA: 1911]  [Article Influence: 273.0]  [Reference Citation Analysis (0)]
40.  Franks I. Barrett esophagus: New insights into the stem cell organization of Barrett esophagus. Nat Rev Gastroenterol Hepatol. 2012;9:125.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
41.  Que J, Garman KS, Souza RF, Spechler SJ. Pathogenesis and Cells of Origin of Barrett's Esophagus. Gastroenterology. 2019;157:349-364.e1.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 86]  [Cited by in RCA: 88]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
42.  Arnold KM, Opdenaker LM, Flynn D, Sims-Mourtada J. Wound healing and cancer stem cells: inflammation as a driver of treatment resistance in breast cancer. Cancer Growth Metastasis. 2015;8:1-13.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 60]  [Cited by in RCA: 86]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
43.  Jenkins GJ, D'Souza FR, Suzen SH, Eltahir ZS, James SA, Parry JM, Griffiths PA, Baxter JN. Deoxycholic acid at neutral and acid pH, is genotoxic to oesophageal cells through the induction of ROS: The potential role of anti-oxidants in Barrett's oesophagus. Carcinogenesis. 2007;28:136-142.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 80]  [Cited by in RCA: 83]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
44.  Shekels LL, Lyftogt CT, Ho SB. Bile acid-induced alterations of mucin production in differentiated human colon cancer cell lines. Int J Biochem Cell Biol. 1996;28:193-201.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 28]  [Cited by in RCA: 33]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
45.  Perez MJ, Briz O. Bile-acid-induced cell injury and protection. World J Gastroenterol. 2009;15:1677-1689.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 469]  [Cited by in RCA: 516]  [Article Influence: 32.3]  [Reference Citation Analysis (3)]
46.  Crowley-Weber CL, Payne CM, Gleason-Guzman M, Watts GS, Futscher B, Waltmire CN, Crowley C, Dvorakova K, Bernstein C, Craven M, Garewal H, Bernstein H. Development and molecular characterization of HCT-116 cell lines resistant to the tumor promoter and multiple stress-inducer, deoxycholate. Carcinogenesis. 2002;23:2063-2080.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 60]  [Cited by in RCA: 67]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
47.  Bomzon A, Holt S, Moore K. Bile acids, oxidative stress, and renal function in biliary obstruction. Semin Nephrol. 1997;17:549-562.  [PubMed]  [DOI]
48.  Ljubuncic P, Fuhrman B, Oiknine J, Aviram M, Bomzon A. Effect of deoxycholic acid and ursodeoxycholic acid on lipid peroxidation in cultured macrophages. Gut. 1996;39:475-478.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 75]  [Cited by in RCA: 90]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
49.  Sokol RJ, Straka MS, Dahl R, Devereaux MW, Yerushalmi B, Gumpricht E, Elkins N, Everson G. Role of oxidant stress in the permeability transition induced in rat hepatic mitochondria by hydrophobic bile acids. Pediatr Res. 2001;49:519-531.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 114]  [Cited by in RCA: 113]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
50.  Mitsuyoshi H, Nakashima T, Sumida Y, Yoh T, Nakajima Y, Ishikawa H, Inaba K, Sakamoto Y, Okanoue T, Kashima K. Ursodeoxycholic acid protects hepatocytes against oxidative injury via induction of antioxidants. Biochem Biophys Res Commun. 1999;263:537-542.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 117]  [Cited by in RCA: 117]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
51.  Shah SA, Volkov Y, Arfin Q, Abdel-Latif MM, Kelleher D. Ursodeoxycholic acid inhibits interleukin 1 beta [corrected] and deoxycholic acid-induced activation of NF-kappaB and AP-1 in human colon cancer cells. Int J Cancer. 2006;118:532-539.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 49]  [Cited by in RCA: 64]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
52.  Huang F. Ursodeoxycholic acid as a potential alternative therapeutic approach for neurodegenerative disorders: Effects on cell apoptosis, oxidative stress and inflammation in the brain. Brain Behav Immun Health. 2021;18:100348.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
53.  Joo SS, Kang HC, Won TJ, Lee DI. Ursodeoxycholic acid inhibits pro-inflammatory repertoires, IL-1 beta and nitric oxide in rat microglia. Arch Pharm Res. 2003;26:1067-1073.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 18]  [Cited by in RCA: 26]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
54.  Miura T, Ouchida R, Yoshikawa N, Okamoto K, Makino Y, Nakamura T, Morimoto C, Makino I, Tanaka H. Functional modulation of the glucocorticoid receptor and suppression of NF-kappaB-dependent transcription by ursodeoxycholic acid. J Biol Chem. 2001;276:47371-47378.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 103]  [Cited by in RCA: 102]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
55.  Yan S, Zhang G, Luo W, Xu M, Peng R, Du Z, Liu Y, Bai Z, Xiao X, Qin S. PROTAC technology: From drug development to probe technology for target deconvolution. Eur J Med Chem. 2024;276:116725.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 20]  [Reference Citation Analysis (0)]