Next Article in Journal
Pig Milk Exosome Packaging ssc-miR-22-3p Alleviates Pig Intestinal Epithelial Cell Injury and Inflammatory Response by Targeting MAPK14
Previous Article in Journal
Metabolic Rate and Oxidative Stress as a Risk Factors in the Development of Colorectal Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Global Transcriptomic Analysis of Topical Sodium Alginate Protection against Peptic Damage in an In Vitro Model of Treatment-Resistant Gastroesophageal Reflux Disease

by
Pelin Ergun
1,
Tina L. Samuels
1,
Angela J. Mathison
2,
Kate Plehhova
3,
Cathal Coyle
3,
Lizzie Horvath
3 and
Nikki Johnston
1,*
1
Department of Otolaryngology and Communication Sciences, Medical College of Wisconsin, Milwaukee, WI 53226, USA
2
Mellowes Center for Genomic Science and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
3
Reckitt Benckiser Healthcare UK Ltd., Slough SL1 3UH, UK
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(19), 10714; https://doi.org/10.3390/ijms251910714
Submission received: 19 September 2024 / Revised: 1 October 2024 / Accepted: 3 October 2024 / Published: 5 October 2024

Abstract

:
Breakthrough symptoms are thought to occur in roughly half of all gastroesophageal reflux disease (GERD) patients despite maximal acid suppression (proton pump inhibitor, PPI) therapy. Topical alginates have recently been shown to enhance mucosal defense against acid-pepsin insult during GERD. We aimed to examine potential alginate protection of transcriptomic changes in a cell culture model of PPI-recalcitrant GERD. Immortalized normal-derived human esophageal epithelial cells underwent pretreatment with commercial alginate-based anti-reflux medications (Gaviscon Advance or Gaviscon Double Action), a matched-viscosity placebo control, or pH 7.4 buffer (sham) alone for 1 min, followed by exposure to pH 6.0 + pepsin or buffer alone for 3 min. RNA sequencing was conducted, and Ingenuity Pathway Analysis was performed with a false discovery rate of ≤0.01 and absolute fold-change of ≥1.3. Pepsin-acid exposure disrupted gene expressions associated with epithelial barrier function, chromatin structure, carcinogenesis, and inflammation. Alginate formulations demonstrated protection by mitigating these changes and promoting extracellular matrix repair, downregulating proto-oncogenes, and enhancing tumor suppressor expression. These data suggest molecular mechanisms by which alginates provide topical protection against injury during weakly acidic reflux and support a potential role for alginates in the prevention of GERD-related carcinogenesis.

1. Introduction

GERD is a chronic public health issue characterized by symptoms of heartburn and/or regurgitation [1]. GERD is a widespread and common disorder [2]. According to research, it affects ~14% of the global population, with significant variability between regions and countries [3]. In a subset of GERD patients, chronic mucosal exposure to gastric contents prompts a metaplastic transformation of the esophageal lining, culminating in Barrett’s esophagus (BE), a precancerous state [4].
Despite the status of PPIs as the first-line therapy for GERD, 50–55% of GERD patients have symptoms that are resistant to PPI treatment [5,6]. PPIs are strong acid-suppressive drugs, yet they do not offer protection against non-acid substances like bile acids and pepsin, and patients frequently continue to have weakly to non-acidic reflux [6,7,8]. Further, long-term PPI use has been associated with negative effects, including bone fractures, kidney disease, dementia, and gastric neoplasia [9,10]. As a result, patients with GERD have become increasingly likely to decline long-term PPI therapy in lieu of alternative solutions [11,12]. While anti-reflux surgery effectively provides symptom relief [13], candidate selection is limited to patients with pathological GERD (particularly severe erosive esophagitis) [12], and fundoplication failure is not uncommon [6].
Natural seaweed-derived alginates have been shown to inhibit pepsin and sequester bile [14] and have long been used as a therapy for mild and moderate GERD. Preclinical and clinical investigations have established their capacity to prevent reflux-attributed mucosal injury and symptoms [15,16,17,18,19,20]. Alginate-antacid anti-reflux medications create a floating barrier (raft) on top of the stomach contents. This raft eliminates or displaces the post-meal “acid pocket”, thus reducing acidic reflux events [21]. Recent work supports an additional role for commercial alginate-antacid solutions [Gaviscon Advance (GA) and Gaviscon Double Action (GDA)] in topical mucosal protection against injury by refluxate constituents such as pepsin, acid, and bile [15,18,22,23]. Given their capacity for comprehensive protection against acid and nonacid reflux constituents, alginates may be a useful adjunctive therapy for patients experiencing breakthrough symptoms despite maximal acid suppression [24]. While the molecular events leading to mucosal damage during GERD have been the focus of extensive research, esophageal damage caused by weakly acidified pepsin, representative of the reflux of patients with PPI-recalcitrant GERD, is poorly understood. Both the molecular pathways of injury elicited by weakly acidified pepsin in the esophagus and the capacity for alginate to rescue such changes warrant further research.
To better understand the role of sodium alginate in topical mucosal protection against molecular injury during PPI-recalcitrant GERD, we investigated whether alginate could shield against transcriptomic alterations caused by weakly acidified pepsin in normal human esophageal cells.

2. Results

Transcriptomic sequencing resulted in a mean total reads per sample of 36,676,200, with a mean of 15,073,461 read gene counts and 77,258,617 read exon counts per sample. Quality control indicated high-quality sequencing data. In principal component analysis (PCA), one sample from GDA+ PA (pepsin-acid) was observed to be significantly different from others and excluded. DE genes were analyzed according to a data-driven approach. Using all detected genes, principal component 1 (PC1) accounted for 65.8% of all gene expression variance, PC2 for 16.7%, and PC3 for 4.5% (Figure 1). The top 100 contributing genes in PC1-3 from PCA analysis of immortalized, normal-derived human esophageal epithelial cells (Het-1A) cells are shown in Supporting Table S1. A heatmap of the conditions and replicates in pairwise comparison are shown in Supporting Figure S1. One thousand and twenty-seven differentially expressed (DE) genes were defined as those having a false discovery rate (FDR) of ≤0.01 and an absolute fold-change (FC) ≥ 1.3 (Supporting Table S2).
Complete names of all gene symbols below are provided in Appendix A. Based on Ingenuity Pathway Analysis (IPA) of DE genes, the top canonical pathways included the Keratinization, Collagen Degradation, and Wound Healing Signaling Pathways, and the top disease/disorders included Gastrointestinal Disease, Organismal Injury and Abnormalities, and Cancer (Table 1, Supporting Table S3, p ≤ 0.005). The top molecular and cellular functions affected by PA were Cell Morphology, Cellular Development, Gene Expression, Cellular Growth and Proliferation, and Cellular Assembly Organization (Table 1 and Supporting Table S3). Top upstream regulators POU2F3, SPRR5, EFNA3, and PAX1 were common across the comparisons of Sham + PA vs. Sham + Sham, Placebo + PA vs. Sham + Sham, and Placebo + PA vs. Sham + PA (Table 1). In the comparison of Gaviscon formulations (GA and GDA) + PA to Sham + PA and Placebo + PA treatments, top upstream regulators included those involved in posttranslational modification SMYD3, HIPK1, SPOP, BRAF, Firre, and miR-3648; signaling molecules TLR5, IL2RG, IL4, and BRAF; and transcription factors HNRNPK and POU2F3 (Table 1).
IPA Tox Function analysis identified biological mechanisms related to toxicity at molecular, cellular, and biochemical levels. Tox Function Pathways were downregulated in these comparisons, as evidenced by the -log (p-value) ≥ 1.3. The analysis revealed that Transforming Growth Factor Beta (TGF-β) Signaling Pathway inhibition was observed in cells treated with PA following GA or GDA pretreatment compared to both Sham + PA and Placebo + PA groups (Figure 2 and Supporting Figure S2) (FC ≥ 1.3). Mechanisms involving Gene Regulation by Peroxisome Proliferation via the Peroxisome proliferator-activated receptor α (PPARα) Pathway were decreased in GA + PA vs. Sham + PA, GDA + PA vs. Sham + PA, and GA + PA vs. Placebo + PA (Figure 2 and Supporting Figure S2).
Ranked by FDR, the top ten up- and downregulated DE genes for each comparison are listed in Table 2 and Supporting Table S4. MT-ND6, FAT4, and LPP were commonly upregulated genes in both GA + PA and GDA + PA groups relative to Sham + PA or Placebo + PA. MZT2A, PCSK1N, FBXL15, and SCAND1 were commonly downregulated genes in both Gaviscon treatments relative to Sham + PA and Placebo + PA. Proto-oncogenic JUN was significantly decreased in GDA + PA versus both Placebo + PA and Sham + PA groups. Placebo + PA decreased FAT4, FAT1, and LPP genes relative to Sham + Sham treatment and decreased the cilia structure gene DNAAF4 and ion channel activity gene TRPC3 relative to Sham + PA. Relative to Sham + Sham, PA treatment upregulated SMOX and AGER and downregulated KRT14, KRT13, and FAT4 genes.

3. Discussion

In untreated GERD patients, refluxate is predominantly acidic (63% pH < 4); however, a significant percentage of reflux events are weakly to non-acidic [25,26]. While PPIs reduce the acidity of refluxate, they do not reduce the frequency of reflux events [27]. Continued weakly or non-acidic reflux is thought to contribute to persistent symptoms exhibited by a large proportion of GERD patients despite PPI therapy [5,6,7,26,28]. Nonacid reflux constituents are also suspected to underly reflux-attributed esophageal carcinogenesis, as indicated by the rising incidence of esophageal adenocarcinoma despite widespread use of PPIs and the association between long-term high adherence to a PPI regimen and greater risk of carcinogenesis and high-grade dysplasia [29]. All gastric juice and refluxate contains pepsin, irrespective of PPI use [8,30]. Evidence demonstrating pepsin’s role in reflux-attributed diseases has developed significantly in recent decades, particularly where it relates to the airways [8,31,32,33]. It is well known that although pepsin’s enzymatic activity decreases as pH levels rise, the enzyme remains stable up to a pH of 8 [34]. While pepsin is enzymatically inactive at neutral pH, it is taken up by airway and esophageal epithelial cells through receptor-mediated endocytosis and is stored in intracellular vesicles of pH 4–5 where it would be reactivated [35,36]. We have previously demonstrated molecular mechanisms of injury by nonacid pepsin in esophageal cells that are consistent with inflammatory and carcinogenic processes attributed to GERD [31,37,38]. The erosive and carcinogenic effects of pepsin can be attenuated by pepsin-inhibiting compounds [15,18,32,39]. Most in vitro research investigating molecular injury during GERD to date has focused on acid alone, or in some cases acidified pepsin [30,40]. Little to no research has been conducted to investigate potential esophageal injury by weakly acidified pepsin. Scientific inquiry to investigate mucosal injury by refluxate pH > 4 is critical to improve our understanding of PPI-recalcitrant GERD.
Clinical studies have shown that combined alginate-antacid formulations can relieve GERD symptoms and provide mucosal protection [41,42,43]. The antacid in these formulations neutralizes acid while the alginate inhibits the enzymatic activity of pepsin and sequesters bile salts [16]. Recent work indicates that alginate formulations provide topical mucosal protection [15,18,22,23]. However, preclinical studies are necessary to better understand how topical alginate may protect against direct and indirect injury to the esophagus caused by gastric contents [44,45,46,47]. Here, we have utilized RNA sequencing to identify molecular pathways altered by pepsin and weak acid in a cell culture model of PPI-recalcitrant GERD and their potential rescue by GA and GDA pretreatment.

3.1. Genes Differentially Expressed by PA Treatment

Our findings indicate that pepsin combined with weak acid caused collagen degradation and keratinization, as indicated by prominent canonical pathways in IPA. KRT13, KRT14, small proline-rich proteins (SPRRs), COL17A1, and MMP-10 were decreased in PA-treated cells relative to sham (Table 2) in accordance with similar findings in the current literature [48,49]. Previous studies have shown that pepsin and weak acid causes depletion of transcripts that encode structural or extracellular matrix components [31,50,51].
In accord with previous in vitro findings [31,32,38,52], PA treatment yielded significant changes in genes with roles in chronic inflammation and cancer development (Table 2). SMOX oxidizes spermine to spermidine and generates reactive oxygen species that cause oxidative-stress-induced apoptosis, increase DNA damage, and promote tumorigenesis [53,54,55,56,57,58]. A non-specific multi-ligand receptor, AGER (RAGE), is involved in cellular cascade responses leading to inflammation both in vitro and in vivo and is implicated in the pathophysiology of various diseases [59,60]. Along with its pro-inflammatory role, AGER overexpression promotes migration, invasion, and epithelial–mesenchymal transition (EMT), features of cancer cells, through ERK signaling [61]. Studies indicate that AGER is overexpressed in various types of malignant tumors, including esophageal and gastric cancers [62,63,64]. Meanwhile, the downregulation of notable tumor suppressors such as FAT4 [65] and CRNN [66] supports the cancerous characteristics of pepsin. The loss or mutations of both genes is related to tumorigenesis in esophageal cells [67,68,69].
PA-induced DE genes also included numerous non-protein coding transcripts that are poorly understood or completely uncharacterized. Mining these findings could yield important insights into novel mechanisms of peptic injury during GERD.

3.2. Alginate Rescue of PA-Induced DE Genes Relative to Placebo or Sham

Collagen degradation was the top canonical pathway attributed to PA treatment; dysregulation of this pathway was rescued by GA but not GDA or placebo (Table 1). Collagen degradation and collagen synthesis (including trimerization) are parts of the extracellular matrix (ECM) remodeling process involved in repair and rebuilding of the ECM after reflux-attributed injury [70,71,72].
Tox function analysis revealed that alginate formulations similarly inhibited injury-associated pathways, such as TGF-β signaling and peroxisome proliferation, via PPARα (Figure 2 and Supporting Figure S2). TGF-β is a key regulatory cytokine involved in numerous cellular processes, including chronic inflammation, proliferation, differentiation, and ECM remodeling [73,74]. Its altered activity contributes to the development of tissue remodeling and progression to malignancy, such as during development of gastrointestinal cancers [74,75], leading to clinical trials investigating inhibition of TGF-β signaling [75,76,77]. Interestingly, TGF-β is overexpressed in the metaplastic stages of esophageal cancer but not in non-neoplastic precancerous cells, such as Barrett’s esophagus (BE) [74,78,79]. However, prolonged inflammatory response may induce the carcinogenic effect of canonical TGF-β signaling in BE or ulcerative colitis [80]. The inhibition of this canonical pathway by alginate relative to sham or non-alginate placebo suggests that alginate may impede the carcinogenic potential of pepsin [31,32,38,52,81,82].
Peroxisome proliferation via the PPARα pathway was the most prominently affected cellular pathway according to IPA Tox Function analysis in GA + PA vs. Sham + PA, GDA + PA vs. Sham + PA, and GA + PA vs. Placebo + PA comparisons (Figure 2 and Supporting Figure S2). The pathway plays several common roles in lipid metabolism, energy homeostasis, inflammation regulation, and cellular detoxification [83] and is involved in different types of cancer progression in complex ways [84,85]. PPAR-mediated transcription occurs when a member of the RXR family interacts with a peroxisome proliferator response element within target genes, triggering transcriptional activation or repression [86]. The Liver X Receptor-Retinoid X Receptor (LXR/RXR) activation pathway was significantly decreased in the GA + PA comparison relative to the Placebo + PA comparison. This pathway plays a role in the regulation of lipid metabolism, cell energy, inflammation, cell detoxification, and various types of cancer [87,88,89].
Our study also showed that GDA pretreatment inhibited the Liver Steatosis Pathway compared to the placebo (GDA + PA vs. Placebo + PA) (Supporting Figure S2). Steatotic liver disease involves having excess fat in the liver and causes chronic inflammation [90]. A recent fatty liver rat model study showed that sodium alginates recover the liver by modulating gut microbiota and suppressing inflammation via the TLR4/NF-κB/NLRP3 inflammatory pathway [91]. Another mice study revealed that alginates prevent liver inflammation and lipid accumulation in the liver. Furthermore, the upregulation of zonula occludens-1 expression showed that intestinal barrier function was impaired despite the low absorption of alginates by the gastrointestinal tract [92]. These pathway analyses demonstrate the inefficacy of the placebo in providing anti-inflammatory and anti-cancer support, highlighting the protective effects of alginate rather than the solution’s viscosity, as supported by the literature [15,18,23,93]. GDA pretreatment also decreased the Mitochondrial Dysfunction Pathway compared to the Sham + PA group (Figure 2). The results support sodium alginate’s homeostatic effects on cell viability [15].
Alginate formulations upregulated tumor suppressors, cell adhesion, signal transduction, regulatory genes, and non-protein coding genes (Table 2, Supporting Table S4). FAT4 (GA + PA vs. Sham + PA, GDA + PA vs. Sham + PA, and GDA + PA vs. Placebo + PA) and FAT1 (GA + PA vs. Sham + PA and GA + PA vs. Placebo + PA) were the top increased tumor suppressors based on FDR. Both are members of the cadherin superfamily and function as adhesion molecules and signaling receptors [94,95]. Many studies have shown the tumor-suppressing role of FAT1 in esophageal cancer cell lines and esophageal squamous cell carcinoma (ESCC) tissues [94,96,97,98]. Furthermore, lower levels of circular FAT1 have been suggested as a novel prognostic marker in ESCC patients [99]. FAT4 is suppressed in various cancer types such as esophageal, gastric, breast, colorectal, liver, and adrenocortical cancer [65,95,100,101]. These two genes were significantly decreased by the direct effect of pepsin and acid treatment compared to untreated cells (Table 2). Pepsin-acid treatment following Gaviscon pretreatments also indicated upregulation of other tumor suppressors, including BRCA2, KLF10, SPRY4, HERC1 (also increased in GDA + PA vs. Sham + PA), SHANK2, RP11, and AHNAK, relative to Placebo + PA or Sham + PA (Table 2, Supporting Table S4).
Alginate formulations downregulated two important proto-oncogenic genes, JUN (GDA + PA vs. Placebo + PA or Sham + PA) and JUNB (GA + PA vs. Placebo + PA) (Table 2, Supporting Table S4). Proto-oncogenes are crucial genes involved in cell growth, differentiation, and survival. They encode proteins that regulate cell division and signal transduction pathways. Several different oncogenes are derived from proto-oncogenes [102]. Overexpression of JUN is detected in patients with laryngeal squamous cell carcinoma and could be related to progression due to its overactivation with c-Fos [103]. Additionally, JUN has inflammatory roles when epithelial cells are exposed to gastric fluids. ERK/c-Jun signaling has been found to regulate the expression of MMP-7 and the degradation of E-cadherin in human pharyngeal epithelial cells [104]. JUNB, a member of the JUN family, is associated with poor prognosis in human gastric cancer and prostate cancer cell lines [105,106]. Beyond its role in cancer, overexpression of JUNB may contribute to various inflammatory diseases [107,108].
Although there is limited research, the loss of MZT2A expression in alginate formulations relative to both placebo and sham PA groups suggests the importance of this gene in rescue of peptic injury. MZT2A controls cell septation signaling during cell development by balancing microtubule polymerization and depolymerization at the centrosome [109]. It is primarily studied in non–small-cell lung cancer, where it is highly expressed in cancer cells compared to normal bronchial cells [109]. Its paralog MZT2B (~96% identity) [110], is overexpressed in patients with gastric cancer [111]. Gaviscon formulations also revealed the downregulation of various genes such as COL6A2 [112], DOHH [113], MEX3D [114], SBNO2 [115], which have roles in cancer or chronic disease progression (Table 2, Supporting Table S4). Further research is needed to better understand the potential role of these mechanisms in reflux-attributed esophageal injury.
Wound healing is a complex, well-coordinated process that includes three phases: inflammation, proliferation, and remodeling. To ensure proper healing following an injury, these processes must be strictly managed. Any dysregulation can lead to incomplete healing, chronic inflammation, excessive tissue damage, and even malignancy [116,117]. While PTGS2 (also known as cyclooxygenase-2, COX-2) is primarily known for its pro-inflammatory roles, it is also a well-known mucosal repair gene [118,119]. Studies have revealed that COX-2 signaling is essential for gastrointestinal epithelial healing. Inhibition of COX-2 can delay gastric ulcer healing and cause significant damage [120,121]. In our study, pepsin and pH 6.0 acid treatment following GA elevated PTGS2 expression relative to Placebo + PA treatment (Supporting Table S4).
Similar to the results of Tox function and Canonical pathway analysis, the top downregulated genes were consistent between comparisons of GDA + PA with both Sham + PA and Placebo + PA (Table 1 and Table 2, Figure 2, Supporting Tables S2–S4). Further, the Placebo + PA versus Sham + PA comparison demonstrated elevation of certain oncogenes (MMP1 [122], NOTCH2NL [123], LIPH [124], and HBQ1 [125]). These results demonstrate the inefficacy of placebo relative to alginate.

3.3. Limitations and Summary

In summary, our study investigates the molecular impact of pepsin and weak acid on normal esophageal epithelial cells and evaluates the protective effects of Gaviscon formulations (GA and GDA) against pepsin-induced dysregulation in GERD-related signaling processes. Using RNA sequencing, we identified significant gene expression changes and pathways associated with inflammation, apoptosis, ECM remodeling, and carcinogenesis. Pepsin exposure notably increased the expression of oncogenic or pro-inflammatory genes while downregulating key tumor suppressors. These data support pepsin’s direct deleterious effect on epithelial barrier integrity and chromatin stability and promotion of a cancer-associated phenotype [15,30,38,126]. Gaviscon formulations demonstrated protective effects by mitigating these detrimental changes, promoting ECM repair, downregulating proto-oncogenes, and enhancing tumor suppressor gene expression. Gaviscon formulations abrogated peptic stimulation of these cancer-associated pathways. These findings provide credence to existing clinical and preclinical evidence that topical sodium alginate protects the esophagus. GDA pretreatment was predominantly associated with RNA regulation and cell signaling, whereas rescue by GA was associated with cell barrier, cell junctional, and lipid metabolism processes. One could surmise that differences between the benefit offered by the different formulations may be related to differences in sodium alginate content; GA has greater sodium alginate content than GDA. While the two formulations demonstrated differences in their ability to rescue certain PA-affected pathways and gene expressions, both formulations conferred benefit relative to placebo, as indicated by comparisons between Gaviscon treatment groups and placebo or sham groups, which demonstrated minimal differences in Tox function analysis, top canonical pathways, top upstream regulators, and significant genes. Our dataset additionally revealed substantial dysregulation of long noncoding RNAs and miRNAs that were dysregulated by PA and rescued by alginate, whose functions in peptic injury of the esophagus warrant further investigation.
There are some limitations to this study. Our dataset includes a single cell culture line (Het-1A) and pH (6.0). Different epithelial cell lines may yield different results and may not adequately represent the clinical situation. Additionally, this model does not account for the potential effects of Helicobacter pylori infection, which is known to exacerbate GERD symptoms and may influence mucosal integrity and inflammation. There is emerging evidence that sodium alginate could have a beneficial effect on H. pylori, a bacterium associated with gastritis and peptic ulcer disease, potentially reducing its impact on GERD progression. The exclusion of H. pylori from the model limits our understanding of how alginate formulations might interact with both GERD and H. pylori-related pathology. Moreover, the study reflects only a descriptive analysis, and no functional validation of the transcriptomic outcomes has been performed. Despite these limitations, the molecular mechanisms identified in this in vitro model may have important implications for GERD patients. Pepsin’s ability to dysregulate oncogenic, inflammatory, and ECM-related pathways suggests a potential role in driving disease progression in acid-suppressed patients with persistent weak acid reflux. The protective effects of Gaviscon formulations observed in vitro may translate into clinically relevant benefits by reducing epithelial damage and possibly lowering the risk of progression to more severe conditions, such as Barrett’s esophagus or esophageal adenocarcinoma. Further clinical studies are warranted to validate these protective effects in human populations. The work herein should be interpreted with this in mind, and results warrant corroboration in vivo and in clinical specimens. Despite these limitations, these data provide preclinical evidence that Gaviscon formulations may provide protection against esophageal injury and carcinogenesis that may be promoted by persistent mucosal exposure to weakly acidified pepsin in acid-suppressed GERD patients.

4. Materials and Methods

4.1. Cell Culture

Immortalized, normal-derived human esophageal epithelial cells (Het-1A) (American Tissue Culture Center, Manassas, VA, USA) were cultured in BEGM (Bronchial Epithelial Cell Growth Medium) medium (Sigma Aldrich, St. Louis, MO, USA). The medium contained the provided supplements of hydrocortisone, insulin, and transferrin, along with 180 μM adenine, 10 ng/mL cholera toxin (both from Sigma-Aldrich), 70 μg/mL bovine pituitary extract, 5% fetal bovine serum, and 1x Antibiotic-Antimycotic (ThermoFisher Scientific, Waltham, MA, USA). The cells were grown on collagen-I coated flasks (Biocoat; Corning, Corning, NY, USA). Once the cells reached 75% confluency, they were subjected to pretreatment conditions in triplicate wells.
The test substances included Gaviscon Advance® (Reckitt Benckiser Group, Slough, UK; containing 1000 mg sodium alginate and 200 mg potassium hydrogen carbonate per 10 mL), labeled as “GA”, and Gaviscon Double Action® (Reckitt Benckiser Group; containing 500 mg sodium alginate, 213 mg sodium bicarbonate, and 325 mg calcium carbonate per 10 mL), labeled as “GDA”. The placebo was a xanthan gum solution with matching viscosity but without alginate or bicarbonates [23]. Pepsin and pH 6.0 treatments were labeled “PA”.
The cultures were pretreated for 1 min in HBSS (Sham) (pH7.4; control, ThermoFisher Scientific, Waltham, MA, USA) or 1:20 dilutions of GA, GDA, and the viscosity-matched placebo control in HBSS. Following pretreatment, the cultures were washed twice with HBSS to mimic the physiologic conditions of esophageal clearance and treated in HBSS pH 7.4 or HBSS pH 6.0 ± 0.1 mg/mL porcine pepsin (Sigma-Aldrich, St. Louis, MO, USA) at 37 °C/5% CO2 for 3 min. After treatment, the cells were rinsed twice with HBSS and allowed to recover in normal growth media under the same conditions for 1 h before harvesting for total RNA extraction. Total RNA was extracted using the RNEasy Plus Mini Kit (Qiagen, Hilden, Germany) which eliminates genomic DNA and QIAshredder columns (Qiagen, Hilden, Germany). RNA quality was assessed by UV spectroscopy (Nanodrop 2000; ThermoFisher, Scientific, Waltham, MA, USA), fluorimetry (Qubit; Thermo Fisher Scientific, Scientific, Waltham, MA, USA), and a fragment analyzer (Agilent, Santa Clara, CA, USA) using high-sensitivity RNA components (Figure 3).

4.2. RNA Sequencing and Pathway Analysis

RNA libraries were prepared (Illumina TruSeq Stranded mRNA, dual indexed, (Illumina, San Diego, CA, USA) and sequenced on the Illumina Nova Seq 6000 (Illumina, San Diego, CA, USA) with 100 bp paired end reads at the Genomic Sciences and Precision Medicine Center (RRID:SCR_022926), as previously described [126]. Briefly, quality control checks were performed using FastQC (v0.11.9, Babraham Bioinformatics, Babraham Institute, Cambridge, UK) and RSeQC (v3.0.0, developed by Dr. Liguo Wang, Mayo Clinic—Rochester, NY, USA). The MAPRSeq3 suite (http://bioinformaticstools.mayo.edu/research/maprseq/, accessed on 1 October 2024) was utilized for analyzing paired-end RNA-Seq data: read alignment was conducted with Star (v2.5.2b, developed by Dr. Alexander Dobin, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA), BAM files were processed using featureCounts (v1.5.1, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia, and exon quantification was obtained via BEDTools (v2, developed by Quinlan Lab, University of Utah, Salt Lake City, UT, USA). This process resulted in gene and exon counts, both raw and normalized for sequencing depth and gene length (linear reads per kilobase of transcript per million). Differential expression (DE) analysis was executed using a pairwise approach with EdgeR (developed by the Walter and Eliza Hall Institute of Medical Research, Parkville, Australia), employing the following thresholds: a minimum of one read per million in at least three samples, a false discovery rate (FDR) of ≤0.01, and an absolute FC ≥ 1.3 [log2(FC) ≥ 0.3785]. Further analysis of differentially expressed genes was conducted using Ingenuity Pathway Analysis (IPA; Qiagen, Hilden, Germany) and principal component analysis (PCA).

5. Conclusions

Our study reveals the molecular impact of pepsin and weak acid on normal esophageal epithelial cells and evaluates the protective effects of sodium alginates against pepsin-induced dysregulation in GERD-related signaling pathways. RNA sequencing identified significant gene expression changes associated with inflammation, apoptosis, ECM remodeling, and carcinogenesis. Pepsin exposure notably increased oncogenic and pro-inflammatory genes while downregulating tumor suppressors, highlighting its detrimental effects on epithelial barrier integrity and chromatin stability. Gaviscon formulations mitigated these harmful changes, promoting ECM repair, downregulating proto-oncogenes, and enhancing tumor suppressor gene expression. The protective effects were attributed to sodium alginate’s mucoadhesive, pepsin-inhibiting, and acid-neutralizing properties, as evidenced by the relative inefficacy of viscosity-matched placebo. Our findings support the role of sodium alginate in protecting the esophagus during PPI-recalcitrant GERD and emphasize the need for further research to explore new therapeutic approaches for this patient population.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms251910714/s1.

Author Contributions

Conceptualization, N.J., T.L.S., K.P. and C.C.; methodology, T.L.S. and N.J.; software, A.J.M. and P.E.; validation, A.J.M., P.E. and T.L.S.; formal analysis, A.J.M. and P.E.; investigation, P.E.; resources, N.J., K.P., L.H. and C.C.; data curation, P.E. and T.L.S.; writing—original draft preparation, P.E.; writing—review and editing, T.L.S., N.J., A.J.M., K.P., L.H. and C.C.; visualization, P.E.; supervision, T.L.S. and N.J.; project administration, N.J.; funding acquisition, N.J. All authors have read and agreed to the published version of the manuscript.

Funding

This study was performed with an unrestricted, investigator-initiated research grant from Reckitt Benckiser UK (Grant Number: FP00023560).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original data presented in the study are openly available in https://zenodo.org/records/13864634 at DOI: 10.5281/zenodo.13864634 (Accessed on 1 October 2024).

Conflicts of Interest

K.P., C.C., and L.H. are employees of Reckitt Benckiser; other authors have no financial relationships or conflict of interest to disclose. This study was presented 23–26 April 2024, at the OESO—16th World Conference Esophagus in Strasbourg, France, and was awarded the Distinguished Abstract Award.

Appendix A

ACAP1 (ArfGAP With Coiled-Coil, Ankyrin Repeat And PH Domains 1), AGER (Advanced Glycosylation End-Product Specific Receptor), AHNAK (Neuroblast differentiation-associated protein), AL121899.2 (AL121899.2), A1BG (Alpha-1-B Glycoprotein), ATP6C (ATP Synthase F0 Complex Subunit C), BEST1 (Bestrophin 1), BRAF (B-Raf Proto-Oncogene, Serine/Threonine Kinase), BRCA2 (BRCA2 DNA Repair Associated), C19orf81 (Chromosome 19 Open Reading Frame 81), CCDC85A (Coiled-Coil Domain Containing 85A), CEMIP (Cell Migration Inducing Hyaluronidase 1), CLEC11A (C-Type Lectin Domain Family 11 Member A), COL6A2 (Collagen Type VI Alpha 2 Chain), CSMD1 (CUB And Sushi Multiple Domains 1), DNAAF4 (Dynein Axonemal Assembly Factor 4), DNHD1 (Dynein Heavy Chain Domain 1), DOHH (Deoxyhypusine Hydroxylase), EEA1 (Early Endosome Antigen 1), EFNA3 (Ephrin A3), EFNA4 (Ephrin A4), EMP1 (Epithelial Membrane Protein 1), EXPH5 (Exophilin 5), FAT1 (FAT Atypical Cadherin 1), FAT4 (FAT Atypical Cadherin 4), FBXL15 (F-Box And Leucine Rich Repeat Protein 15), FGD6 (FYVE, RhoGEF And PH Domain Containing 6), FIRRE (Functional Intergenic Repeating RNA Element), FSCN3 (Fascin Actin-Bundling Protein 3), GARIN1A (Gamma-Aminobutyric Acid Receptor Interacting Protein Alpha 1), GCNT4 (Glucosaminyl (N-Acetyl) Transferase 4), GPR50 (G Protein-Coupled Receptor 50), GRIN3B (Glutamate Ionotropic Receptor NMDA Type Subunit 3B), HERC1 (HECT And RLD Domain Containing E3 Ubiquitin Protein Ligase 1), HES4 (Hes Family BHLH Transcription Factor 4), HNRNPK (Heterogeneous Nuclear Ribonucleoprotein K), IL2RG (Interleukin 2 Receptor Subunit Gamma), ITK (IL2 Inducible T-Cell Kinase), IGSF1 (Immunoglobulin Superfamily Member 1), JUN (Jun Proto-Oncogene, AP-1 Transcription Factor Subunit), KLF2 (Kruppel-Like Factor 2), KLF10 (Kruppel-Like Factor 10), KRT13 (Keratin 13), KRT14 (Keratin 14), LPP (Lipoma Preferred Partner), MMP1 (Matrix Metallopeptidase 1), MZT2A (Mitotic Spindle Organizing Protein 2A), MYCBP2 (MYC Binding Protein 2), NICOL1 (Neuronal Intracellular Coiled-Coil Protein 1), NOTCH2NLA (Notch 2 N-Terminal Like A), NOTCH2NLB (Notch 2 N-Terminal Like B), NUTM2E (NUT Midline Carcinoma Family Member 2E), PADI1 (Peptidyl Arginine Deiminase 1), PAX1 (Paired Box 1), PCSK1N (Proprotein Convertase Subtilisin/Kexin Type 1 Inhibitor), PDE3A (Phosphodiesterase 3A), PDZRN3 (PDZ Domain Containing Ring Finger 3), PGAP1 (Post-GPI Attachment To Proteins 1), POLR2J2/POLR2J3 (RNA Polymerase II Subunit J2/J3), PRAM1 (PML-RAR Alpha Regulated Adaptor Molecule 1), PRR7 (Proline Rich 7), RAPGEF2 (Rap Guanine Nucleotide Exchange Factor 2), RAMP1 (Receptor Activity Modifying Protein 1), RP11 (Retinitis Pigmentosa Type 11), SBNO2 (Strawberry Notch Homolog 2), SCAND1 (SCAND1 N-Terminal Domain Containing 1), SHANK2 (SH3 And Multiple Ankyrin Repeat Domains 2), ST8SIA4 (ST8 Alpha-N-Acetyl-Neuraminide Alpha-2,8-Sialyltransferase 4), SMOX (Spermine Oxidase), SMYD3 (SET And MYND Domain Containing 3), SNX22 (Sorting Nexin 22), SPOP (Speckle-Type POZ Protein), SPRR2A (Small Proline Rich Protein 2A), SPRR5 (Small Proline Rich Protein 5), TLR5 (Toll Like Receptor 5), TMEM91 (Transmembrane Protein 91), TPGS1 (Tropomyosin Receptor Kinase Fused Gene), TRIM2 (Tripartite Motif Containing 2), TTP2A (Tetratricopeptide Repeat Domain 2A), UBR4 (Ubiquitin Protein Ligase E3 Component N-Recognin 4), ZBTB20 (Zinc Finger And BTB Domain Containing 20), ZNF837 (Zinc Finger Protein 837).

References

  1. Zerbib, F.; Bredenoord, A.J.; Fass, R.; Kahrilas, P.J.; Roman, S.; Savarino, E.; Sifrim, D.; Vaezi, M.; Yadlapati, R.; Gyawali, C.P. ESNM/ANMS consensus paper: Diagnosis and management of refractory gastro-esophageal reflux disease. Neurogastroenterol. Motil. 2021, 33, e14075. [Google Scholar] [CrossRef]
  2. Yadlapati, R.; Gyawali, C.P.; Pandolfino, J.E.; Participants, C.G.C.C. AGA Clinical Practice Update on the Personalized Approach to the Evaluation and Management of GERD: Expert Review. Clin. Gastroenterol. Hepatol. 2022, 20, 984–994.e981. [Google Scholar] [CrossRef]
  3. Nirwan, J.S.; Hasan, S.S.; Babar, Z.U.; Conway, B.R.; Ghori, M.U. Global Prevalence and Risk Factors of Gastro-oesophageal Reflux Disease (GORD): Systematic Review with Meta-analysis. Sci. Rep. 2020, 10, 5814. [Google Scholar] [CrossRef] [PubMed]
  4. Ergun, P.; Kipcak, S.; Bor, S. Epigenetic Alterations from Barrett’s Esophagus to Esophageal Adenocarcinoma. Int. J. Mol. Sci. 2023, 24, 7817. [Google Scholar] [CrossRef]
  5. Delshad, S.D.; Almario, C.V.; Chey, W.D.; Spiegel, B.M.R. Prevalence of Gastroesophageal Reflux Disease and Proton Pump Inhibitor-Refractory Symptoms. Gastroenterology 2020, 158, 1250–1261.e1252. [Google Scholar] [CrossRef] [PubMed]
  6. Davis, T.A.; Gyawali, C.P. Refractory Gastroesophageal Reflux Disease: Diagnosis and Management. J. Neurogastroenterol. Motil. 2024, 30, 17–28. [Google Scholar] [CrossRef] [PubMed]
  7. Patel, D.; Fass, R.; Vaezi, M. Untangling Nonerosive Reflux Disease From Functional Heartburn. Clin. Gastroenterol. Hepatol. 2021, 19, 1314–1326. [Google Scholar] [CrossRef]
  8. Ergun, P.; Kipcak, S.; Dettmar, P.W.; Fisher, J.; Woodcock, A.D.; Bor, S. Pepsin and pH of Gastric Juice in Patients with Gastrointestinal Reflux Disease and Subgroups. J. Clin. Gastroenterol. 2022, 56, 512–517. [Google Scholar] [CrossRef]
  9. Castellana, C.; Pecere, S.; Furnari, M.; Telese, A.; Matteo, M.V.; Haidry, R.; Eusebi, L.H. Side effects of long-term use of proton pump inhibitors: Practical considerations. Pol. Arch. Intern. Med. 2021, 131, 541–549. [Google Scholar] [CrossRef]
  10. Fossmark, R.; Martinsen, T.C.; Waldum, H.L. Adverse Effects of Proton Pump Inhibitors-Evidence and Plausibility. Int. J. Mol. Sci. 2019, 20, 5203. [Google Scholar] [CrossRef]
  11. Ergun, P.; Kipcak, S.; Selvi Gunel, N.; Yildirim Sozmen, E.; Bor, S. Inflammatory responses in esophageal mucosa before and after laparoscopic antireflux surgery. World J. Gastrointest. Surg. 2024, 16, 871–881. [Google Scholar] [CrossRef]
  12. Pauwels, A.; Boecxstaens, V.; Andrews, C.N.; Attwood, S.E.; Berrisford, R.; Bisschops, R.; Boeckxstaens, G.E.; Bor, S.; Bredenoord, A.J.; Cicala, M.; et al. How to select patients for antireflux surgery? The ICARUS guidelines (international consensus regarding preoperative examinations and clinical characteristics assessment to select adult patients for antireflux surgery). Gut 2019, 68, 1928–1941. [Google Scholar] [CrossRef] [PubMed]
  13. Pessaux, P.; Arnaud, J.P.; Delattre, J.F.; Meyer, C.; Baulieux, J.; Mosnier, H. Laparoscopic antireflux surgery: Five-year results and beyond in 1340 patients. Arch Surg 2005, 140, 946–951. [Google Scholar] [CrossRef] [PubMed]
  14. Leiman, D.A.; Riff, B.P.; Morgan, S.; Metz, D.C.; Falk, G.W.; French, B.; Umscheid, C.A.; Lewis, J.D. Alginate therapy is effective treatment for GERD symptoms: A systematic review and meta-analysis. Dis. Esophagus 2017, 30, 1–9. [Google Scholar] [CrossRef]
  15. Samuels, T.L.; Blaine-Sauer, S.; Yan, K.; Plehhova, K.; Coyle, C.; Johnston, N. Topical Alginate Protection against Pepsin-Mediated Esophageal Damage: E-Cadherin Proteolysis and Matrix Metalloproteinase Induction. Int. J. Mol. Sci. 2023, 24. [Google Scholar] [CrossRef]
  16. Strugala, V.; Avis, J.; Jolliffe, I.G.; Johnstone, L.M.; Dettmar, P.W. The role of an alginate suspension on pepsin and bile acids—Key aggressors in the gastric refluxate. Does this have implications for the treatment of gastro-oesophageal reflux disease? J. Pharm. Pharmacol. 2009, 61, 1021–1028. [Google Scholar] [CrossRef]
  17. Sweis, R.; Kaufman, E.; Anggiansah, A.; Wong, T.; Dettmar, P.; Fried, M.; Schwizer, W.; Avvari, R.K.; Pal, A.; Fox, M. Post-prandial reflux suppression by a raft-forming alginate (Gaviscon Advance) compared to a simple antacid documented by magnetic resonance imaging and pH-impedance monitoring: Mechanistic assessment in healthy volunteers and randomised, controlled, double-blind study in reflux patients. Aliment. Pharmacol. Ther. 2013, 37, 1093–1102. [Google Scholar] [CrossRef]
  18. Samuels, T.L.; Yan, K.; Patel, N.; Plehhova, K.; Coyle, C.; Hurley, B.P.; Johnston, N. Alginates for Protection Against Pepsin-Acid Induced Aerodigestive Epithelial Barrier Disruption. Laryngoscope 2022, 132, 2327–2334. [Google Scholar] [CrossRef]
  19. Giannini, E.G.; Zentilin, P.; Dulbecco, P.; Iiritano, E.; Bilardi, C.; Savarino, E.; Mansi, C.; Savarino, V. A comparison between sodium alginate and magaldrate anhydrous in the treatment of patients with gastroesophageal reflux symptoms. Dig. Dis. Sci. 2006, 51, 1904–1909. [Google Scholar] [CrossRef]
  20. Wilkinson, J.; Wade, A.; Thomas, S.J.; Jenner, B.; Hodgkinson, V.; Coyle, C. Randomized clinical trial: A double-blind, placebo-controlled study to assess the clinical efficacy and safety of alginate-antacid (Gaviscon Double Action) chewable tablets in patients with gastro-oesophageal reflux disease. Eur. J. Gastroenterol. Hepatol. 2019, 31, 86–93. [Google Scholar] [CrossRef]
  21. Rohof, W.O.; Bennink, R.J.; Smout, A.J.; Thomas, E.; Boeckxstaens, G.E. An alginate-antacid formulation localizes to the acid pocket to reduce acid reflux in patients with gastroesophageal reflux disease. Clin. Gastroenterol. Hepatol. 2013, 11, 1585–1591; quiz e1590. [Google Scholar] [CrossRef]
  22. Sonmez, S.; Coyle, C.; Sifrim, D.; Woodland, P. Duration of adhesion of swallowed alginates to distal oesophageal mucosa: Implications for topical therapy of oesophageal diseases. Aliment. Pharmacol. Ther. 2020, 52, 442–448. [Google Scholar] [CrossRef] [PubMed]
  23. Woodland, P.; Batista-Lima, F.; Lee, C.; Preston, S.L.; Dettmar, P.; Sifrim, D. Topical protection of human esophageal mucosal integrity. Am. J. Physiol. Gastrointest. Liver Physiol. 2015, 308, G975–G980. [Google Scholar] [CrossRef]
  24. Ranaldo, N.; Losurdo, G.; Iannone, A.; Principi, M.; Barone, M.; De Carne, M.; Ierardi, E.; Di Leo, A. Tailored therapy guided by multichannel intraluminal impedance pH monitoring for refractory non-erosive reflux disease. Cell Death Dis. 2017, 8, e3040. [Google Scholar] [CrossRef] [PubMed]
  25. Sadatomi, D.; Kono, T.; Mogami, S.; Fujitsuka, N. Weak acids induce PGE(2) production in human oesophageal cells: Novel mechanisms underlying GERD symptoms. Sci. Rep. 2020, 10, 20775. [Google Scholar] [CrossRef] [PubMed]
  26. Namasivayam, V.; Arora, A.S.; Murray, J.A. Weakly acidic reflux. Dis. Esophagus 2011, 24, 56–62. [Google Scholar] [CrossRef]
  27. Tamhankar, A.P.; Peters, J.H.; Portale, G.; Hsieh, C.C.; Hagen, J.A.; Bremner, C.G.; DeMeester, T.R. Omeprazole does not reduce gastroesophageal reflux: New insights using multichannel intraluminal impedance technology. J. Gastrointest. Surg. 2004, 8, 890–897; discussion 897–898. [Google Scholar] [CrossRef]
  28. Zikos, T.A.; Clarke, J.O. Non-acid Reflux: When It Matters and Approach to Management. Curr. Gastroenterol. Rep. 2020, 22, 43. [Google Scholar] [CrossRef]
  29. Hvid-Jensen, F.; Pedersen, L.; Funch-Jensen, P.; Drewes, A.M. Proton pump inhibitor use may not prevent high-grade dysplasia and oesophageal adenocarcinoma in Barrett’s oesophagus: A nationwide study of 9883 patients. Aliment Pharmacol Ther 2014, 39, 984–991. [Google Scholar] [CrossRef]
  30. Sharma, P.; Yadlapati, R. Pathophysiology and treatment options for gastroesophageal reflux disease: Looking beyond acid. Ann. N. Y Acad. Sci. 2021, 1486, 3–14. [Google Scholar] [CrossRef]
  31. Samuels, T.L.; Altman, K.W.; Gould, J.C.; Kindel, T.; Bosler, M.; MacKinnon, A.; Hagen, C.E.; Johnston, N. Esophageal pepsin and proton pump synthesis in barrett’s esophagus and esophageal adenocarcinoma. Laryngoscope 2019, 129, 2687–2695. [Google Scholar] [CrossRef] [PubMed]
  32. Blaine-Sauer, S.; Samuels, T.L.; Yan, K.; Johnston, N. The Protease Inhibitor Amprenavir Protects against Pepsin-Induced Esophageal Epithelial Barrier Disruption and Cancer-Associated Changes. Int. J. Mol. Sci. 2023, 24, 6765. [Google Scholar] [CrossRef] [PubMed]
  33. Lechien, J.R.; Bobin, F. Variability and accuracy of multiple saliva pepsin measurements in laryngopharyngeal reflux patients. J. Otolaryngol. Head. Neck Surg. 2023, 52, 66. [Google Scholar] [CrossRef]
  34. Piper, D.W.; Fenton, B.H. pH stability and activity curves of pepsin with special reference to their clinical importance. Gut 1965, 6, 506–508. [Google Scholar] [CrossRef]
  35. Johnston, N.; Dettmar, P.W.; Bishwokarma, B.; Lively, M.O.; Koufman, J.A. Activity/stability of human pepsin: Implications for reflux attributed laryngeal disease. Laryngoscope 2007, 117, 1036–1039. [Google Scholar] [CrossRef]
  36. Johnston, N.; Wells, C.W.; Samuels, T.L.; Blumin, J.H. Rationale for targeting pepsin in the treatment of reflux disease. Ann. Otol. Rhinol. Laryngol. 2010, 119, 547–558. [Google Scholar] [CrossRef] [PubMed]
  37. Samuels, T.; Hoekzema, C.; Gould, J.; Goldblatt, M.; Frelich, M.; Bosler, M.; Lee, S.H.; Johnston, N. Local Synthesis of Pepsin in Barrett’s Esophagus and the Role of Pepsin in Esophageal Adenocarcinoma. Ann. Otol. Rhinol. Laryngol. 2015, 124, 893–902. [Google Scholar] [CrossRef]
  38. Stabenau, K.A.; Samuels, T.L.; Lam, T.K.; Mathison, A.J.; Wells, C.; Altman, K.W.; Battle, M.A.; Johnston, N. Pepsinogen/Proton Pump Co-Expression in Barrett’s Esophageal Cells Induces Cancer-Associated Changes. Laryngoscope 2023, 133, 59–69. [Google Scholar] [CrossRef] [PubMed]
  39. Nagahama, K.; Yamato, M.; Nishio, H.; Takeuchi, K. Essential role of pepsin in pathogenesis of acid reflux esophagitis in rats. Dig. Dis. Sci. 2006, 51, 303–309. [Google Scholar] [CrossRef]
  40. Ergun, P.; Capanoglu, D.; Kipcak, S.; Bor, S. Response of Esophageal Epithelium to Acute and Chronic Stress in Rabbits. Bull. Exp. Biol. Med. 2021, 171, 582–587. [Google Scholar] [CrossRef]
  41. Rettura, F.; Bronzini, F.; Campigotto, M.; Lambiase, C.; Pancetti, A.; Berti, G.; Marchi, S.; de Bortoli, N.; Zerbib, F.; Savarino, E.; et al. Refractory Gastroesophageal Reflux Disease: A Management Update. Front Med. 2021, 8, 765061. [Google Scholar] [CrossRef] [PubMed]
  42. Reimer, C.; Lodrup, A.B.; Smith, G.; Wilkinson, J.; Bytzer, P. Randomised clinical trial: Alginate (Gaviscon Advance) vs. placebo as add-on therapy in reflux patients with inadequate response to a once daily proton pump inhibitor. Aliment. Pharmacol. Ther. 2016, 43, 899–909. [Google Scholar] [CrossRef] [PubMed]
  43. Scarpellini, E.; Ang, D.; Pauwels, A.; De Santis, A.; Vanuytsel, T.; Tack, J. Management of refractory typical GERD symptoms. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 281–294. [Google Scholar] [CrossRef] [PubMed]
  44. Ergun, P.; Kipcak, S.; Gunel, N.S.; Bor, S.; Sozmen, E.Y. Roles of Cytokines in Pathological and Physiological Gastroesophageal Reflux Exposure. J. Neurogastroenterol. Motil. 2023, 30, 290–302. [Google Scholar] [CrossRef] [PubMed]
  45. Souza, R.F.; Huo, X.; Mittal, V.; Schuler, C.M.; Carmack, S.W.; Zhang, H.Y.; Zhang, X.; Yu, C.; Hormi-Carver, K.; Genta, R.M.; et al. Gastroesophageal reflux might cause esophagitis through a cytokine-mediated mechanism rather than caustic acid injury. Gastroenterology 2009, 137, 1776–1784. [Google Scholar] [CrossRef]
  46. Dunbar, K.B.; Agoston, A.T.; Odze, R.D.; Huo, X.; Pham, T.H.; Cipher, D.J.; Castell, D.O.; Genta, R.M.; Souza, R.F.; Spechler, S.J. Association of Acute Gastroesophageal Reflux Disease With Esophageal Histologic Changes. JAMA 2016, 315, 2104–2112. [Google Scholar] [CrossRef]
  47. Orlando, R.C. The integrity of the esophageal mucosa. Balance between offensive and defensive mechanisms. Best. Pr. Res. Clin. Gastroenterol. 2010, 24, 873–882. [Google Scholar] [CrossRef]
  48. Samuels, T.L.; Johnston, N. Pepsin as a causal agent of inflammation during nonacidic reflux. Otolaryngol. Head. Neck Surg. 2009, 141, 559–563. [Google Scholar] [CrossRef]
  49. Tan, J.J.; Dai, Y.F.; Wang, F.; Lv, Z.H.; Huang, L.J.; Peng, L.Y.; Li, X.P. Pepsin-mediated inflammation in laryngopharyngeal reflux via the ROS/NLRP3/IL-1beta signaling pathway. Cytokine 2024, 178, 156568. [Google Scholar] [CrossRef]
  50. McCann, A.J.; Samuels, T.L.; Blumin, J.H.; Johnston, N. The role of pepsin in epithelia-mesenchymal transition in idiopathic subglottic stenosis. Laryngoscope 2020, 130, 154–158. [Google Scholar] [CrossRef]
  51. Korbut, E.; Janmaat, V.T.; Wierdak, M.; Hankus, J.; Wojcik, D.; Surmiak, M.; Magierowska, K.; Brzozowski, T.; Peppelenbosch, M.P.; Magierowski, M. Molecular Profile of Barrett’s Esophagus and Gastroesophageal Reflux Disease in the Development of Translational Physiological and Pharmacological Studies. Int. J. Mol. Sci. 2020, 21, 6436. [Google Scholar] [CrossRef]
  52. Tan, J.J.; Wang, L.; Mo, T.T.; Wang, J.; Wang, M.G.; Li, X.P. Pepsin promotes IL-8 signaling-induced epithelial-mesenchymal transition in laryngeal carcinoma. Cancer Cell Int. 2019, 19, 64. [Google Scholar] [CrossRef] [PubMed]
  53. Fadista, J.; Yakimov, V.; Vosa, U.; Hansen, C.S.; Kasela, S.; Skotte, L.; Geller, F.; Courraud, J.; Esko, T.; Kukuskina, V.; et al. Genetic regulation of spermine oxidase activity and cancer risk: A Mendelian randomization study. Sci. Rep. 2021, 11, 17463. [Google Scholar] [CrossRef] [PubMed]
  54. Kim, S.; Kim, D.; Roh, S.; Hong, I.; Kim, H.; Ahn, T.S.; Kang, D.H.; Lee, M.S.; Baek, M.J.; Kwak, H.J.; et al. Expression of Spermine Oxidase Is Associated with Colorectal Carcinogenesis and Prognosis of Patients. Biomedicines 2022, 10, 626. [Google Scholar] [CrossRef]
  55. Gobert, A.P.; Al-Greene, N.T.; Singh, K.; Coburn, L.A.; Sierra, J.C.; Verriere, T.G.; Luis, P.B.; Schneider, C.; Asim, M.; Allaman, M.M.; et al. Distinct Immunomodulatory Effects of Spermine Oxidase in Colitis Induced by Epithelial Injury or Infection. Front. Immunol. 2018, 9, 1242. [Google Scholar] [CrossRef] [PubMed]
  56. Chaturvedi, R.; Asim, M.; Romero-Gallo, J.; Barry, D.P.; Hoge, S.; de Sablet, T.; Delgado, A.G.; Wroblewski, L.E.; Piazuelo, M.B.; Yan, F.; et al. Spermine oxidase mediates the gastric cancer risk associated with Helicobacter pylori CagA. Gastroenterology 2011, 141, 1696–1708.e2. [Google Scholar] [CrossRef] [PubMed]
  57. Murray-Stewart, T.; Sierra, J.C.; Piazuelo, M.B.; Mera, R.M.; Chaturvedi, R.; Bravo, L.E.; Correa, P.; Schneider, B.G.; Wilson, K.T.; Casero, R.A. Epigenetic silencing of miR-124 prevents spermine oxidase regulation: Implications for Helicobacter pylori-induced gastric cancer. Oncogene 2016, 35, 5480–5488. [Google Scholar] [CrossRef]
  58. Murray Stewart, T.; Dunston, T.T.; Woster, P.M.; Casero, R.A., Jr. Polyamine catabolism and oxidative damage. J. Biol. Chem. 2018, 293, 18736–18745. [Google Scholar] [CrossRef]
  59. Dong, H.; Zhang, Y.; Huang, Y.; Deng, H. Pathophysiology of RAGE in inflammatory diseases. Front. Immunol. 2022, 13, 931473. [Google Scholar] [CrossRef]
  60. Hudson, B.I.; Lippman, M.E. Targeting RAGE Signaling in Inflammatory Disease. Annu. Rev. Med. 2018, 69, 349–364. [Google Scholar] [CrossRef]
  61. Chen, M.C.; Chen, K.C.; Chang, G.C.; Lin, H.; Wu, C.C.; Kao, W.H.; Teng, C.J.; Hsu, S.L.; Yang, T.Y. RAGE acts as an oncogenic role and promotes the metastasis of human lung cancer. Cell Death Dis. 2020, 11, 265. [Google Scholar] [CrossRef] [PubMed]
  62. Jing, R.R.; Cui, M.; Sun, B.L.; Yu, J.; Wang, H.M. Tissue-specific expression profiling of receptor for advanced glycation end products and its soluble forms in esophageal and lung cancer. Genet. Test. Mol. Biomark. 2010, 14, 355–361. [Google Scholar] [CrossRef]
  63. Faruqui, T.; Khan, M.S.; Akhter, Y.; Khan, S.; Rafi, Z.; Saeed, M.; Han, I.; Choi, E.H.; Yadav, D.K. RAGE Inhibitors for Targeted Therapy of Cancer: A Comprehensive Review. Int. J. Mol. Sci. 2022, 24, 266. [Google Scholar] [CrossRef] [PubMed]
  64. Li, L.; Beeraka, N.M.; Xie, L.; Dong, L.; Liu, J.; Wang, L. Co-expression of High-mobility group box 1 protein (HMGB1) and receptor for advanced glycation end products (RAGE) in the prognosis of esophageal squamous cell carcinoma. Discov. Oncol. 2022, 13, 64. [Google Scholar] [CrossRef]
  65. Mao, W.; Zhou, J.; Hu, J.; Zhao, K.; Fu, Z.; Wang, J.; Mao, K. A pan-cancer analysis of FAT atypical cadherin 4 (FAT4) in human tumors. Front. Public Health 2022, 10, 969070. [Google Scholar] [CrossRef] [PubMed]
  66. Hong, F.; Wan, X.; Bai, Y. Identification of Down-Expressed CRNN Associated with Cancer Progression and Poor Prognosis in Laryngeal Squamous Cell Carcinoma. Front Biosci. 2024, 29, 125. [Google Scholar] [CrossRef]
  67. Kaur, R.; Chauhan, A.; Bhat, S.A.; Chatterjee, D.; Ghoshal, S.; Pal, A. Gene of the month: Cornulin. J. Clin. Pathol. 2022, 75, 289–291. [Google Scholar] [CrossRef]
  68. Liu, X.; Zhao, S.; Wang, K.; Zhou, L.; Jiang, M.; Gao, Y.; Yang, R.; Yan, S.; Zhang, W.; Lu, B.; et al. Spatial transcriptomics analysis of esophageal squamous precancerous lesions and their progression to esophageal cancer. Nat. Commun. 2023, 14, 4779. [Google Scholar] [CrossRef]
  69. Visser, E.; Franken, I.A.; Brosens, L.A.A.; de Leng, W.W.J.; Strengman, E.; Offerhaus, J.A.; Ruurda, J.P.; van Hillegersberg, R. Targeted next-generation sequencing of commonly mutated genes in esophageal adenocarcinoma patients with long-term survival. Dis. Esophagus 2017, 30, 1–8. [Google Scholar] [CrossRef]
  70. Onursal, C.; Dick, E.; Angelidis, I.; Schiller, H.B.; Staab-Weijnitz, C.A. Collagen Biosynthesis, Processing, and Maturation in Lung Ageing. Front Med. 2021, 8, 593874. [Google Scholar] [CrossRef]
  71. Abu-Romman, A.; Scholand, K.K.; Govindarajan, G.; Yu, Z.; Pal-Ghosh, S.; Stepp, M.A.; de Paiva, C.S. Age-Related Differences in the Mouse Corneal Epithelial Transcriptome and Their Impact on Corneal Wound Healing. Investig. Ophthalmol. Vis. Sci. 2024, 65, 21. [Google Scholar] [CrossRef] [PubMed]
  72. Deraman, M.A.; Abdul Hafidz, M.I.; Lawenko, R.M.; Ma, Z.F.; Wong, M.S.; Coyle, C.; Lee, Y.Y. Randomised clinical trial: The effectiveness of Gaviscon Advance vs non-alginate antacid in suppression of acid pocket and post-prandial reflux in obese individuals after late-night supper. Aliment. Pharmacol. Ther. 2020, 51, 1014–1021. [Google Scholar] [CrossRef] [PubMed]
  73. Massague, J.; Sheppard, D. TGF-beta signaling in health and disease. Cell 2023, 186, 4007–4037. [Google Scholar] [CrossRef] [PubMed]
  74. Luo, J.; Chen, X.Q.; Li, P. The Role of TGF-beta and Its Receptors in Gastrointestinal Cancers. Transl. Oncol. 2019, 12, 475–484. [Google Scholar] [CrossRef] [PubMed]
  75. Gough, N.R.; Xiang, X.; Mishra, L. TGF-beta Signaling in Liver, Pancreas, and Gastrointestinal Diseases and Cancer. Gastroenterology 2021, 161, 434–452.e415. [Google Scholar] [CrossRef]
  76. Zhang, M.; Zhang, Y.Y.; Chen, Y.; Wang, J.; Wang, Q.; Lu, H. TGF-beta Signaling and Resistance to Cancer Therapy. Front. Cell Dev. Biol. 2021, 9, 786728. [Google Scholar] [CrossRef]
  77. Ali, S.; Rehman, M.U.; Yatoo, A.M.; Arafah, A.; Khan, A.; Rashid, S.; Majid, S.; Ali, A.; Ali, M.N. TGF-beta signaling pathway: Therapeutic targeting and potential for anti-cancer immunity. Eur. J. Pharmacol. 2023, 947, 175678. [Google Scholar] [CrossRef]
  78. Lee, S.W.; Lien, H.C.; Lin, C.C.; Wen, M.C.; Chang, C.S. Low Expression of Transforming Growth Factor beta in the Epithelium of Barrett’s Esophagus. Gastroenterol. Res. 2018, 11, 189–194. [Google Scholar] [CrossRef]
  79. Chen, Y.; Zhu, S.; Liu, T.; Zhang, S.; Lu, J.; Fan, W.; Lin, L.; Xiang, T.; Yang, J.; Zhao, X.; et al. Epithelial cells activate fibroblasts to promote esophageal cancer development. Cancer Cell 2023, 41, 903–918.e908. [Google Scholar] [CrossRef]
  80. Tambunting, L.; Kelleher, D.; Duggan, S.P. The Immune Underpinnings of Barrett’s-Associated Adenocarcinogenesis: A Retrial of Nefarious Immunologic Co-Conspirators. Cell Mol. Gastroenterol. Hepatol. 2022, 13, 1297–1315. [Google Scholar] [CrossRef]
  81. Doukas, P.G.; Vageli, D.P.; Sasaki, C.T.; Judson, B.L. Pepsin Promotes Activation of Epidermal Growth Factor Receptor and Downstream Oncogenic Pathways, at Slightly Acidic and Neutral pH, in Exposed Hypopharyngeal Cells. Int. J. Mol. Sci. 2021, 22, 4275. [Google Scholar] [CrossRef] [PubMed]
  82. Yin, C.Y.; Zhang, S.S.; Zhong, J.T.; Zhou, S.H. Pepsin and Laryngeal and Hypopharyngeal Carcinomas. Clin. Exp. Otorhinolaryngol. 2021, 14, 159–168. [Google Scholar] [CrossRef]
  83. Dai, J.; Li, Y.; Kametani, F.; Cui, X.; Igarashi, Y.; Huo, J.; Miyahara, H.; Mori, M.; Higuchi, K. Curcumin promotes AApoAII amyloidosis and peroxisome proliferation in mice by activating the PPARalpha signaling pathway. Elife 2021, 10, 63538. [Google Scholar] [CrossRef]
  84. Wang, Y.; Lei, F.; Lin, Y.; Han, Y.; Yang, L.; Tan, H. Peroxisome proliferator-activated receptors as therapeutic target for cancer. J. Cell Mol. Med. 2023, 28, e17931. [Google Scholar] [CrossRef] [PubMed]
  85. Wagner, N.; Wagner, K.D. The Role of PPARs in Disease. Cells 2020, 9, 2367. [Google Scholar] [CrossRef] [PubMed]
  86. Nickkho-Amiry, M.; McVey, R.; Holland, C. Peroxisome proliferator-activated receptors modulate proliferation and angiogenesis in human endometrial carcinoma. Mol. Cancer Res. 2012, 10, 441–453. [Google Scholar] [CrossRef]
  87. Zhu, M.; Li, Y.; Ding, C.; Wang, J.; Ma, Y.; Li, Z.; Zhang, X.; Wang, P. Proteomic Profiling Change and Its Implies in the Early Mycosis Fungoides (MF) Using Isobaric Tags for Relative and Absolute Quantification (iTRAQ). Biomed. Res. Int. 2020, 2020, 9237381. [Google Scholar] [CrossRef]
  88. Wong, Y.L.; Ramanathan, A.; Yuen, K.M.; Mustafa, W.M.W.; Abraham, M.T.; Tay, K.K.; Rahman, Z.A.A.; Chen, Y. Comparative sera proteomics analysis of differentially expressed proteins in oral squamous cell carcinoma. PeerJ 2021, 9, e11548. [Google Scholar] [CrossRef]
  89. Si, X.; Xu, F.; Xu, F.; Wei, M.; Ge, Y.; Chenge, S. CADM1 inhibits ovarian cancer cell proliferation and migration by potentially regulating the PI3K/Akt/mTOR pathway. Biomed. Pharmacother. 2020, 123, 109717. [Google Scholar] [CrossRef]
  90. Paternostro, R.; Trauner, M. Current treatment of non-alcoholic fatty liver disease. J. Intern. Med. 2022, 292, 190–204. [Google Scholar] [CrossRef]
  91. Zhao, H.; Gao, X.; Liu, Z.; Zhang, L.; Fang, X.; Sun, J.; Zhang, Z.; Sun, Y. Sodium Alginate Prevents Non-Alcoholic Fatty Liver Disease by Modulating the Gut-Liver Axis in High-Fat Diet-Fed Rats. Nutrients 2022, 14, 4846. [Google Scholar] [CrossRef] [PubMed]
  92. Kawauchi, S.; Horibe, S.; Sasaki, N.; Tanahashi, T.; Mizuno, S.; Hamaguchi, T.; Rikitake, Y. Inhibitory Effects of Sodium Alginate on Hepatic Steatosis in Mice Induced by a Methionine- and Choline-deficient Diet. Mar. Drugs 2019, 17, 104. [Google Scholar] [CrossRef]
  93. Woodland, P.; Lee, C.; Duraisamy, Y.; Farre, R.; Dettmar, P.; Sifrim, D. Assessment and protection of esophageal mucosal integrity in patients with heartburn without esophagitis. Am. J. Gastroenterol. 2013, 108, 535–543. [Google Scholar] [CrossRef] [PubMed]
  94. Zhai, Y.; Shan, C.; Zhang, H.; Kong, P.; Zhang, L.; Wang, Y.; Hu, X.; Cheng, X. FAT1 downregulation enhances stemness and cisplatin resistance in esophageal squamous cell carcinoma. Mol. Cell Biochem. 2022, 477, 2689–2702. [Google Scholar] [CrossRef]
  95. Huang, F.Y.; Wong, D.K.; Mak, L.Y.; Cheung, T.T.; Zhang, S.S.; Chau, H.T.; Hui, R.W.; Seto, W.K.; Yuen, M.F. FAT4 loss initiates hepatocarcinogenesis through the switching of canonical to noncanonical WNT signaling pathways. Hepatol. Commun. 2023, 7, e0338. [Google Scholar] [CrossRef]
  96. Lu, Y.; Wang, Z.; Zhou, L.; Ma, Z.; Zhang, J.; Wu, Y.; Shao, Y.; Yang, Y. FAT1 and PTPN14 Regulate the Malignant Progression and Chemotherapy Resistance of Esophageal Cancer through the Hippo Signaling Pathway. Anal. Cell Pathol. 2021, 2021, 9290372. [Google Scholar] [CrossRef]
  97. Wang, Y.; Wang, G.; Ma, Y.; Teng, J.; Wang, Y.; Cui, Y.; Dong, Y.; Shao, S.; Zhan, Q.; Liu, X. FAT1, a direct transcriptional target of E2F1, suppresses cell proliferation, migration and invasion in esophageal squamous cell carcinoma. Chin. J. Cancer Res. 2019, 31, 609–619. [Google Scholar] [CrossRef] [PubMed]
  98. Yang, B.; Zhang, Y.K.; Ye, Y.J.; Liu, T.D.; Peng, S.Z.; Zhang, R.S. Fat1 inhibits cell proliferation via ERK signaling pathway in esophageal squamous cell carcinoma. Zhonghua Zhong Liu Za Zhi 2021, 43, 523–527. [Google Scholar] [CrossRef]
  99. Takaki, W.; Konishi, H.; Shoda, K.; Arita, T.; Kataoka, S.; Shibamoto, J.; Furuke, H.; Takabatake, K.; Shimizu, H.; Komatsu, S.; et al. Significance of Circular FAT1 as a Prognostic Factor and Tumor Suppressor for Esophageal Squamous Cell Carcinoma. Ann. Surg. Oncol. 2021, 28, 8508–8518. [Google Scholar] [CrossRef]
  100. Wang, L.; Li, K.; Wang, C.; Shi, X.; Yang, H. miR-107 regulates growth and metastasis of gastric cancer cells via activation of the PI3K-AKT signaling pathway by down-regulating FAT4. Cancer Med. 2019, 8, 5264–5273. [Google Scholar] [CrossRef]
  101. Wu, H.; Yu, Z.; Liu, Y.; Guo, L.; Teng, L.; Guo, L.; Liang, L.; Wang, J.; Gao, J.; Li, R.; et al. Genomic characterization reveals distinct mutation landscapes and therapeutic implications in neuroendocrine carcinomas of the gastrointestinal tract. Cancer Commun. 2022, 42, 1367–1386. [Google Scholar] [CrossRef] [PubMed]
  102. Kontomanolis, E.N.; Koutras, A.; Syllaios, A.; Schizas, D.; Mastoraki, A.; Garmpis, N.; Diakosavvas, M.; Angelou, K.; Tsatsaris, G.; Pagkalos, A.; et al. Role of Oncogenes and Tumor-suppressor Genes in Carcinogenesis: A Review. Anticancer. Res. 2020, 40, 6009–6015. [Google Scholar] [CrossRef] [PubMed]
  103. Tsiambas, E.; Mastronikolis, N.; P, P.F.; Kyrodimos, E.; Chrysovergis, A.; Papanikolaou, V.; Mastronikolis, S.; Peschos, D.; Roukas, D.; Ragos, V. c-Jun/c-Fos complex in laryngeal squamous cell carcinoma. J. BUON 2020, 25, 618–620. [Google Scholar]
  104. Im, N.R.; Kim, B.; Jung, K.Y.; Baek, S.K. Matrix metalloproteinase-7 induces E-cadherin cleavage in acid-exposed primary human pharyngeal epithelial cells via the ROS/ERK/c-Jun pathway. J. Mol. Med. 2022, 100, 313–322. [Google Scholar] [CrossRef]
  105. Li, S.; Wei, Y.; Sun, X.; Liu, M.; Zhu, M.; Yuan, Y.; Zhang, J.; Dong, Y.; Hu, K.; Ma, S.; et al. JUNB mediates oxaliplatin resistance via the MAPK signaling pathway in gastric cancer by chromatin accessibility and transcriptomic analysis. Acta Biochim. Biophys. Sin. 2023, 55, 1784–1796. [Google Scholar] [CrossRef]
  106. Roumeliotou, A.; Strati, A.; Chamchougia, F.; Xagara, A.; Tserpeli, V.; Smilkou, S.; Lagopodi, E.; Christopoulou, A.; Kontopodis, E.; Drositis, I.; et al. Comprehensive Analysis of CXCR4, JUNB, and PD-L1 Expression in Circulating Tumor Cells (CTCs) from Prostate Cancer Patients. Cells 2024, 13, 782. [Google Scholar] [CrossRef]
  107. Ren, F.J.; Cai, X.Y.; Yao, Y.; Fang, G.Y. JunB: A paradigm for Jun family in immune response and cancer. Front. Cell Infect. Microbiol. 2023, 13, 1222265. [Google Scholar] [CrossRef]
  108. Wang, S.; Wang, Y.; Han, B.; Chen, Y.; Bai, X.; Yu, S.; Liu, M. Huanglian ointment alleviates eczema by maintaining the balance of c-Jun and JunB and inhibiting AGE-RAGE-mediated pro-inflammation signaling pathway. Phytomedicine 2022, 105, 154372. [Google Scholar] [CrossRef]
  109. Wang, H.; Jiang, X.; Cheng, Y.; Ren, H.; Hu, Y.; Zhang, Y.; Su, H.; Zou, Z.; Wang, Q.; Liu, Z.; et al. MZT2A promotes NSCLC viability and invasion by increasing Akt phosphorylation via the MOZART2 domain. Cancer Sci. 2021, 112, 2210–2222. [Google Scholar] [CrossRef]
  110. Wieczorek, M.; Huang, T.L.; Urnavicius, L.; Hsia, K.C.; Kapoor, T.M. MZT Proteins Form Multi-Faceted Structural Modules in the gamma-Tubulin Ring Complex. Cell Rep. 2020, 31, 107791. [Google Scholar] [CrossRef]
  111. Heitor da Silva Maues, J.; Ferreira Ribeiro, H.; de Maria Maues Sacramento, R.; Maia de Sousa, R.; Pereira de Tommaso, R.; Dourado Kovacs Machado Costa, B.; Cardoso Soares, P.; Pimentel Assumpcao, P.; de Fatima Aquino Moreira-Nunes, C.; Mario Rodriguez Burbano, R. Downregulated genes by silencing MYC pathway identified with RNA-SEQ analysis as potential prognostic biomarkers in gastric adenocarcinoma. Aging 2020, 12, 24651–24670. [Google Scholar] [CrossRef] [PubMed]
  112. Zhu, J.; Lin, Q.; Zheng, H.; Rao, Y.; Ji, T. The pro-invasive factor COL6A2 serves as a novel prognostic marker of glioma. Front. Oncol. 2022, 12, 897042. [Google Scholar] [CrossRef] [PubMed]
  113. Guo, Q.; Zhang, Y.C.; Wang, W.; Wang, Y.Q.; Liu, Y.; Yang, Z.; Zhao, M.M.; Feng, N.; Wang, Y.H.; Zhang, X.W.; et al. Deoxyhypusine hydroxylase as a novel pharmacological target for ischemic stroke via inducing a unique post-translational hypusination modification. Pharmacol. Res. 2022, 176, 106046. [Google Scholar] [CrossRef] [PubMed]
  114. Shao, L.; Wang, J.; Karatas, O.; Ittmann, M. MEX3D is an oncogenic driver in prostate cancer. Prostate 2021, 81, 1202–1213. [Google Scholar] [CrossRef]
  115. Wu, R.; Sun, J.Y.; Zhao, L.L.; Fan, Z.N.; Yang, C. Systematic Identification of Key Functional Modules and Genes in Gastric Cancer. Biomed. Res. Int. 2020, 2020, 8853348. [Google Scholar] [CrossRef]
  116. Gilman, K.E.; Limesand, K.H. The complex role of prostaglandin E(2)-EP receptor signaling in wound healing. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2021, 320, R287–R296. [Google Scholar] [CrossRef]
  117. Greten, F.R.; Grivennikov, S.I. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019, 51, 27–41. [Google Scholar] [CrossRef]
  118. Cheng, H.; Huang, H.; Guo, Z.; Chang, Y.; Li, Z. Role of prostaglandin E2 in tissue repair and regeneration. Theranostics 2021, 11, 8836–8854. [Google Scholar] [CrossRef] [PubMed]
  119. Zheng, C.; Qu, Y.X.; Wang, B.; Shen, P.F.; Xu, J.D.; Chen, Y.X. COX-2/PGE2 facilitates fracture healing by activating the Wnt/beta-catenin signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 9721–9728. [Google Scholar] [CrossRef]
  120. Haworth, R.; Oakley, K.; McCormack, N.; Pilling, A. Differential expression of COX-1 and COX-2 in the gastrointestinal tract of the rat. Toxicol. Pathol. 2005, 33, 239–245. [Google Scholar] [CrossRef]
  121. Wallace, J.L.; Devchand, P.R. Emerging roles for cyclooxygenase-2 in gastrointestinal mucosal defense. Br. J. Pharmacol. 2005, 145, 275–282. [Google Scholar] [CrossRef] [PubMed]
  122. Zhang, J.; Zhang, N.; Yang, X.; Xin, X.; Jia, C.H.; Li, S.; Lu, Q.; Jiang, T.; Wang, T. Machine Learning and Novel Biomarkers Associated with Immune Infiltration for the Diagnosis of Esophageal Squamous Cell Carcinoma. J. Oncol. 2022, 2022, 6732780. [Google Scholar] [CrossRef] [PubMed]
  123. Funato, K.; Smith, R.C.; Saito, Y.; Tabar, V. Dissecting the impact of regional identity and the oncogenic role of human-specific NOTCH2NL in an hESC model of H3.3G34R-mutant glioma. Cell Stem Cell 2021, 28, 894–905.e897. [Google Scholar] [CrossRef] [PubMed]
  124. Gao, P.; Liu, Q.; Ai, B.; Fang, Y.; Wang, Z.; Wang, J. Prognostic Value and Clinical Significance of LIPH in Breast Cancer. Int. J. Gen. Med. 2021, 14, 7613–7623. [Google Scholar] [CrossRef]
  125. Kim, K.; Choi, E.Y.; Ahn, H.M.; Kim, D.G.; Kim, Y.J. Hemoglobin Subunit Theta 1 Promotes Proliferation by Reducing Reactive Oxygen Species in Lung Adenocarcinoma. Cancers 2023, 15, 5504. [Google Scholar] [CrossRef]
  126. Samuels, T.L.; Zimmermann, M.T.; Zeighami, A.; Demos, W.; Southwood, J.E.; Blumin, J.H.; Bock, J.M.; Johnston, N. RNA Sequencing Reveals Cancer-Associated Changes in Laryngeal Cells Exposed to Non-Acid Pepsin. Laryngoscope 2021, 131, 121–129. [Google Scholar] [CrossRef]
Figure 1. PCA of RNA sequence data of Het-1A cells treated with Gaviscon Advance + PA (light blue), Gaviscon Double Action (dark blue), a matched placebo control + PA (light green), pH 7.0 buffer (sham) (dark green), and sham + PA (pink). PC1 accounted for 65.8% of the variance, PC2 for 16.7%, and PC3 for 4.5%. PCA = principal component analysis; GA = Gaviscon Advance; GDA = Gaviscon Double Action; PA = Pepsin + acid, Het-1A = immortalized, normal-derived human esophageal epithelial cells.
Figure 1. PCA of RNA sequence data of Het-1A cells treated with Gaviscon Advance + PA (light blue), Gaviscon Double Action (dark blue), a matched placebo control + PA (light green), pH 7.0 buffer (sham) (dark green), and sham + PA (pink). PC1 accounted for 65.8% of the variance, PC2 for 16.7%, and PC3 for 4.5%. PCA = principal component analysis; GA = Gaviscon Advance; GDA = Gaviscon Double Action; PA = Pepsin + acid, Het-1A = immortalized, normal-derived human esophageal epithelial cells.
Ijms 25 10714 g001
Figure 2. IPA Tox Function analysis. Dark blue: GA + PA vs. Sham + PA. Blue: GDA + PA vs. Sham + PA. Blue color indicates z-score is negative. GA = Gaviscon Advance; GDA = Gaviscon Double Action; PA = pepsin + acid.
Figure 2. IPA Tox Function analysis. Dark blue: GA + PA vs. Sham + PA. Blue: GDA + PA vs. Sham + PA. Blue color indicates z-score is negative. GA = Gaviscon Advance; GDA = Gaviscon Double Action; PA = pepsin + acid.
Ijms 25 10714 g002
Figure 3. Research workflow. Sham: pH7.4 HBSS, Placebo: xanthan gum solution with matching viscosity but without alginate or bicarbonates, GA = Gaviscon Advance; GDA = Gaviscon Double Action; PA = pepsin + acid.
Figure 3. Research workflow. Sham: pH7.4 HBSS, Placebo: xanthan gum solution with matching viscosity but without alginate or bicarbonates, GA = Gaviscon Advance; GDA = Gaviscon Double Action; PA = pepsin + acid.
Ijms 25 10714 g003
Table 1. Ingenuity Pathway Analysis of significant DE transcripts.
Table 1. Ingenuity Pathway Analysis of significant DE transcripts.
Sham + PA vs. Sham + Sham
Top Canonical Pathwaysp-ValueOverlap
Keratinization7.01 × 10−72.8% 6/214
Collagen Degradation3.74 × 10−33.3% 2/61
Melatonin Degradation II5.95 × 10−325.0% 1/4
Top Diseasesp-Value RangeNumber of Molecules
Gastrointestinal Disease4.78 × 10−2–2.28 × 10−833
Organismal Injury and Abnormalities4.95 × 10−2–2.28 × 10−836
Cancer4.73 × 10−2–4.19 × 10−536
Molecular and Cellular Functionsp-Value Range#Number of Molecules
Cell Morphology 4.95 × 10−2–2.67 × 10−511
Cellular Development 4.80 × 10−2–1.46 × 10−421
Cellular Growth and Proliferation4.80 × 10−2–1.46 × 10−420
Cell Death and Survival4.89 × 10−2–4.08 × 10−418
Cellular Assembly and Organization4.66 × 10−2–4.45 × 10−414
Top Networks Score
Gastrointestinal Disease, Organismal Injury and Abnormalities, Cardiovascular System,
Development and Function
56
Cancer, Organismal Injury and Abnormalities, Cell Death and Survival24
Top Upstream Regulatorsp-Value Range
POU2F35.91 × 10−6
SPRR52.95 × 10−5
EFNA33.38 × 10−5
PAX13.86 × 10−5
USP124.94 × 10−5
GA + PA vs. Sham + PA
Top Canonical Pathwaysp-ValueOverlap
Polyamine Regulation in Colon Cancer4.95 × 10−45.1% 3/59
Collagen Degradation5.45 × 10−44.9% 3/61
Collagen Chain Trimerization5.90 × 10−34.5% 2/44
Top Diseasesp-Value RangeNumber of Molecules
Organismal Injury and Abnormalities4.98 × 10−2–9.96 × 10−560
Developmental Disorders4.34 × 10−2–3.62 × 10−415
Connective Tissue Disorders4.84 × 10−2–3.62 × 10−411
Molecular and Cellular Functionsp-Value RangeNumber of Molecules
Cell Death and Survival4.98 × 10−2–2.61 × 10−312
Cell-To-Cell Signaling and Interaction3.63 × 10−2–2.61 × 10−36
Cellular Development4.59 × 10−2–2.61 × 10−312
Cellular Growth and Proliferation4.59 × 10−2–2.61 × 10−38
Cellular Movement4.84 × 10−2–2.61 × 10−38
Top Networks Score
Cardiovascular System, Development and Function, Organismal Development, Cardiovascular
Disease
53
Cell Death and Survival, Organismal Injury and Abnormalities, Cell Signaling32
Top Upstream Regulatorsp-Value Range
HNRNPK2.73 × 10−4
HIPK13.24 × 10−4
N1,N11-diethylnorspermine7.00 × 10−4
TLR57.22 × 10−4
Salmonella enterica serotype abortus equilibrium polysaccharide8.09 × 10−4
GDA + PA vs. Sham + PA
Top Canonical Pathwaysp-ValueOverlap
Synaptic Adhesion-like Molecules3.53 × 10−414.3% 3/21
TGF- Signaling4.62 × 10−45.2% 5/96
Collagen Degradation7.37 × 10−46.6% 4/61
Top Diseasesp-Value RangeNumber of Molecules
Cancer3.93 × 10−2–1.08 × 10−12156
Endocrine System Disorders3.48 × 10−2–1.08 × 10−12138
Organismal Injury and Abnormalities3.93 × 10−2–1.08 × 10−12157
Molecular and Cellular Functionsp-Value RangeNumber of Molecules
Cellular Assembly and Organization3.93 × 10−2–2.62 × 10−429
Gene Expression3.93 × 10−2–3.67 × 10−441
Cellular Movement3.93 × 10−2–9.06 × 10−416
Cell Death and Survival3.93 × 10−2–1.37 × 10−337
Cellular Development3.93 × 10−2–1.37 × 10−359
Top Networks Score
Cardiovascular System, Development and Function, Cellular Development, Cellular Growth and
Proliferation
35
Cancer, Organismal Injury and Abnormalities, Developmental Disorders35
Top Upstream Regulatorsp-Value Range
SMYD38.21 × 10−6
Firre1.31 × 10−5
SPOP1.86 × 10−5
FMN25.11 × 10−5
IL2RG5.99 × 10−5
Placebo + PA vs. Sham + PA
Top Canonical Pathwaysp-ValueOverlap
Keratinization5.89 × 10−103.3% 7/214
Collagen Degradation1.40 × 10−33.3% 2/61
Wound Healing Signaling1.49 × 10−31.2% 3/252
Top Diseasesp-Value RangeNumber of Molecules
Gastrointestinal Disease2.25 × 10−2–9.24 × 10−109
Organismal Injury and Abnormalities4.89 × 10−2–9.24 × 10−1022
Dermatological Diseases and Conditions4.89 × 10−2–3.77 × 10−710
Molecular and Cellular Functionsp-Value RangeNumber of Molecules
Cell Morphology1.98 × 10−2–5.86 × 10−63
Cellular Movement3.79 × 10−2–1.03 × 10−44
Cellular Development 4.63 × 10−2–8.46 × 10−47
Carbohydrate Metabolism 9.10 × 10−4–9.10 × 10−41
Cell Cycle4.54 × 10−3–9.10 × 10−42
Top Networks Score
Gastrointestinal Disease, Organismal Injury and Abnormalities, Dermatological Diseases and
Conditions
33
Lipid Metabolism, Molecular Transport, Small Molecule Biochemistry23
Top Upstream Regulatorsp-Value Range
POU2F34.10 × 10−6
EFNA46.35 × 10−6
EFNA37.30 × 10−6
PAX17.80 × 10−6
SPRR51.02 × 10−5
Table 2. Top 10 significant differentially expressed transcripts.
Table 2. Top 10 significant differentially expressed transcripts.
Based on False Discovery Rate
UpregulatedDownregulated UpregulatedDownregulated
Sham + PA vs.
Sham + Sham
SAPCD1TPGS1Placebo + PA vs.
Sham + PA
MMP1DNAAF4
BEST1KRT14ZNF837TRPC3
SMOXKRT13CCDC85ATIGD3
AGERFAT4LIPHNUTM2E
TMEM91RAPGEF2SPRR2EGPRIN3
FSCN3POLR2J2PDZRN3PRAM1
RAMP1MIURFNOTCH2NLST8SIA4
SNX22EEA1HBQ1PADI1
SLC52A1HERC1GRIN3BGARIN1A
ACAP1LPPAL121899.2ITK
UpregulatedDownregulated UpregulatedDownregulated
GA + PA vs.
Sham + PA
MT-ND6FBXL15GDA + PA vs.
Sham + PA
MT-ND6FBXL15
FAT4PCSK1NEMP1JUN
RAPGEF2MZT2AFAT4MEX3D
POLR2J2SCAND1GCNT4ATP6C
HERC1TPGS1AHNAKCOL6A2
LPPDOHHPOLR2J2PCSK1N
POLR2J3CLEC11AHERC1MZT2A
RP11CIMAP1BPOLR2J3KLF2
NCR3LG1HES4RP11SBNO2
FAT1PRR7UBR4SCAND1
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ergun, P.; Samuels, T.L.; Mathison, A.J.; Plehhova, K.; Coyle, C.; Horvath, L.; Johnston, N. Global Transcriptomic Analysis of Topical Sodium Alginate Protection against Peptic Damage in an In Vitro Model of Treatment-Resistant Gastroesophageal Reflux Disease. Int. J. Mol. Sci. 2024, 25, 10714. https://doi.org/10.3390/ijms251910714

AMA Style

Ergun P, Samuels TL, Mathison AJ, Plehhova K, Coyle C, Horvath L, Johnston N. Global Transcriptomic Analysis of Topical Sodium Alginate Protection against Peptic Damage in an In Vitro Model of Treatment-Resistant Gastroesophageal Reflux Disease. International Journal of Molecular Sciences. 2024; 25(19):10714. https://doi.org/10.3390/ijms251910714

Chicago/Turabian Style

Ergun, Pelin, Tina L. Samuels, Angela J. Mathison, Kate Plehhova, Cathal Coyle, Lizzie Horvath, and Nikki Johnston. 2024. "Global Transcriptomic Analysis of Topical Sodium Alginate Protection against Peptic Damage in an In Vitro Model of Treatment-Resistant Gastroesophageal Reflux Disease" International Journal of Molecular Sciences 25, no. 19: 10714. https://doi.org/10.3390/ijms251910714

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop