Skip to main content

REVIEW article

Front. Oncol., 22 February 2023
Sec. Cancer Epidemiology and Prevention
This article is part of the Research Topic Perioperative Management and Cancer Outcome View all 11 articles

A potential link between aberrant expression of ECRG4 and atrial fibrillation

  • 1Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
  • 2School of Medicine, Shanghai University, Shanghai, China

Esophageal cancer-related gene-4 (ECRG4), a 148-amino acid propertied and new tumor suppressor, is initially cloned from the normal esophageal epithelium. ECRG4 was found to be expressed not only in esophageal tissues but also in cardiomyocytes. Previous studies demonstrated that ECRG4 is constitutively expressed in esophageal epithelial cells, and its degree of downregulation is directly proportional to prognosis in patients with esophageal cancer. In the heart, ECRG4 shows greater expression in the atria than in the ventricles, which accounts for its heterogeneity. Downregulation of ECRG4 expression level correlates with esophageal cancer, as well as myocardial injuries and arrhythmias. As a result, this review summarizes the possible susceptibility gene, ECRG4 and its associated molecular mechanisms in cancer patients with atrial fibrillation and myocardial injury. The review begins by describing ECRG4’s biological background, discusses its expression in the cardiovascular system, lists the clinical and animal research related to the downregulation of ECRG4 in atrial fibrillation, and focuses on its potential role in atrial fibrillation. Downregulation of ECRG4 may increase the risk of atrial fibrillation by affecting ion channels, MMPs expression and inflammatory response. We will then discuss how ECRG4 can be used in the treatment of tumors and arrhythmias, and provide a novel possible strategy to reduce the occurrence of perioperative cardiovascular adverse events in patients with tumors such as esophageal cancer and gastric cancer.

Introduction

Esophageal cancer-related gene-4 (ECRG4) is a newly identified tumor suppressor gene and a sentinel molecule for maintaining tissue homeostasis. Recent research has revealed that ECRG4 expression is quite distinct and is present in esophageal squamous epithelial cells as well as the sinoatrial node, atrioventricular node, atrial and ventricular cells. Additional investigations further concluded that ECRG4 could maintain cardiac homeostasis and regulate cardiac rhythm while it downregulation may contribute to atrial fibrillation (AF) (1). Moreover, ECRG4 is most likely a hypoxic sensor and may be related to myocardial ischemia (1). Notably, ECRG4 is also a tumor suppressor gene that can prevent esophageal cancer cell proliferation (2). Downregulation of this gene expression increases the risk of esophageal cancer and is strongly linked to a poor patient prognosis (24). Epidemiological studies also suggest a strong correlation between AF and various tumors. For instance, the risk of colorectal cancer in AF patients was ten times greater than in those without AF, according to a case-control study (5). Furthermore, reduced ECRG4 expression in esophageal cancer patients is associated with an increased incidence of myocardial injury and atrial fibrillation (2, 6). Collectively, the above studies show that ECRG4 may be implicated in both tumorigenesis and cardiovascular adverse events. Herein, we describe the ECRG4’s biological background, discuss its expression in the cardiovascular system, list the clinical and animal research related to the downregulation of ECRG4 in atrial fibrillation, and focus on its potential role in atrial fibrillation. We will then discuss how ECRG4 can be used in the treatment of tumors and arrhythmias, and provide a novel possible strategy to reduce the occurrence of perioperative cardiovascular adverse events in patients with tumors such as esophageal cancer and gastric cancer.

The biological background of ECRG4

Su et al. first discovered ECRG4 in normal human esophageal epithelial cells (7), and it was eventually localized in the c2orf40 locus of chromosome 2, which consists of four exons spanning approximately 14.9 kilobases (4, 8). The initial bioinformatic analysis and subsequent biochemical characterization indicate that the protein encoded by ECRG4 is a hormone-like secretory protein. The ECRG4 gene also encodes a protein with a molecular weight of 17KDA, and peptides with different molecular weights associated with the ECRG4 protein were also identified. Most of the tumor suppressor genes are usually intracellular or membrane proteins. Unlike most tumor suppressor genes, which are usually membrane or intracellular proteins, ECRG4 is a 148 amino acid propeptide that is covalently linked to the amino end on cell surfaces (9, 10). Since ECRG4 is attached to the cell surface, it acts as a “sentinel” in maintaining tissue homeostasis (Figure 1). The presence of ECRG4 on the cell surface suggests that its homeostasis is maintained. After tissue injury, ECRG4 can quickly detach from the cell surface (within 24 hours), thereby increasing tissue injury responses like vascular leakage, immune cell infiltration, and cell proliferation (in 2-4 days). The injury response gradually disappears during the healing process as ECRG4 returns to the cellular surface (usually in 6-7 days) (1113).

FIGURE 1
www.frontiersin.org

Figure 1 Potential functions of ECRG4 in the esophagus and cardiovascular system.

Previous studies demonstrate that ECRG4 is constitutively expressed in esophageal epithelial cells, and its degree of downregulation is directly proportional to prognosis in esophageal cancer patients (Figure 1) (2). Studies have shown that ECRG4 may induce the downregulation of COX-2 through the NF-КB pathway, thereby inhibiting tumor growth in esophageal squamous cell carcinoma (ESCC) (2). Other Studies have found that ECRG4 can directly interact with ECRG1 to up-regulate the expression of p21, induce G1 phase arrest of cell cycle, and inhibit the proliferation of cancer cells (3). Further research revealed that ECRG4 was down-regulated to varying degrees in gastric cancer (14, 15), breast cancer (16, 17), hepatocellular cancer (18, 19), nasopharyngeal cancer (2022), laryngeal cancer (23), bladder cancer (24, 25), glioma (26), colorectal cancer and prostate cancer (2628). These findings suggest that ECRG4 plays a tumor-suppressive role. Various other cells/tissues, such as the adrenal gland, choroid plexus, cardiomyocytes, and conduction system, also express ECRG4. ECRG4 is known to regulate inflammation (1113), induce neuronal senescence (29), participate in the formation of atrial fibrillation (30), and possibly act as a hypoxia sensor, contributing to myocardial injury.

ECRG4: Expression in the cardiovascular system

In 2017 Mirabeau et al. uncovered ECRG4 as a new secretory peptide in mouse endocrine tissues as well as in other locations, including the pituitary, adrenal gland, pancreas, choroid plexus, and the atrioventricular node of the heart (8). Notably, ECRG4 mRNAs are expressed in heart (31). It was found that ECRG4 was expressed in the sinoatrial node, atrium and ventricle, and ECRG4 expression was higher in atrium (30, 31). Porzionato et al. used immunohistochemical analysis to show that ECRG4 was uniformly expressed in normal rat atrial myocytes while only expressed in sporadic ventricular myocytes (31). Professor Dang further discovered that down-regulating ECRG4 in atrial myocytes activated pro-inflammatory cascade and genes involved in heart remodeling, which participated in the occurrence of atrial fibrillation, and concluded that the normal expression of ECRG4 could maintain cardiac homeostasis (Figure 1) (30). Other studies have shown that ECRG4 promotes cardiovascular homeostasis and prevents atrial fibrillation by regulating the response to ischemia/hypoxia (32).

ECRG4 downregulation is associated with atrial fibrillation: Clinical and animal research

Atrial fibrillation is the most common sustained cardiac tachyarrhythmia encountered by physicians, with an ever rising incidence globally. Studies have found that ECRG4 is implicated in the pathogenesis of atrial fibrillation (Figure 2). Moreover, since ECRG4 is a tumor suppressor gene, the downregulation of ECRG4 may also be a risk factor for atrial fibrillation in cancer patients. Extensive epidemiological data show a strong correlation between AF incidence and cancer, which has garnered widespread attention. Erichsen found a tenfold increase in the incidence of colorectal cancer in patients with atrial fibrillation (5). A cohort study found that the cancer diagnosis rate in newly diagnosed AF patients increased 5-fold compared to the expected cancer incidence in the general population after a 3-month follow-up (33). Consistently, Conen found that cancer patients experienced twice the incidence of AF during surgery as non-tumor patients (34). Another clinical control study showed that in esophageal cancer patients with intraoperative hypothermia, the incidence of postoperative myocardial injury could rise to 31.4%, while the incidence of atrial fibrillation (AF) was 14.3% (6), significantly higher than the myocardial injury incidence of 8% in other non-cardiac surgeries (35) and 2.9% incidence of atrial fibrillation in thoracoscopic lung cancer surgery (36). Previous studies reported that the incidence of new-onset atrial fibrillation after radical esophagectomy was 12-37% (3747). Atrial fibrillation may also be linked to lower ECRG4 expression in other patients besides tumor patients Five suitable atrial appendage specimens from patients with rheumatic heart disease with or without atrial fibrillation were further collected clinically to investigate the expression of ECRG4 in the heart, and immunohistochemistry confirmed the downregulation of ECRG4 in the atrial appendage of patients with atrial fibrillation (30).

FIGURE 2
www.frontiersin.org

Figure 2 Mechanism of atrial fibrillation induced by down-regulation ECRG4.

Additionally, certain drugs may affect ECRG4 expression and cause atrial fibrillation. Doxorubicin(DOX) is well-known for cardiotoxic effects, including atrial fibrillation. According to a prospective study by Kilickap and colleagues, DOX-containing regimens caused arrhythmia in 19 patients (65.5%) of 29 patients with various cancers, of whom 3 patients (10.3%) had paroxysmal AF (48). In a previous study, Long et al. investigated the role of ECRG4 in AF and myocardial injury induced by DOX (49). DOX decreased endogenous ECRG4 gene expression in the heart and cultured neonatal rat cardiomyocytes. Further, cardiomyocyte-specific conditional ECRG4 knockout mice showed increased sensitivity to DOX-induced cardiotoxicity due to abnormal signaling pathways, including Oxidative phosphorylation, Thermogenesis, Diabetic cardiomyopathy, Cardiac muscle contraction (Figure 3). This study suggests that ECRG4, which is constitutively expressed in the heart, can maintain cardiac homeostasis and protect cardiomyocytes from the cardiac toxicity caused by DOX (49). Taken together, the high incidence of AF in tumor patients may be related to ECRG4 expression. Therefore, it is crucial to investigate how ECRG4 maintains cardiac homeostasis and regulates cardiac rhythm.

FIGURE 3
www.frontiersin.org

Figure 3 ECRG4 is involved in the mechanism of doxorubicin induced myocardial injury. Figure shows the 5’UTR gene sequence of rat ECRG4 -878/+3, which contains 3 potential SP1 transcription factors. Doxorubicin inhibited the expression of SP1 and reduced the binding of SP1 to the rat ECRG4 promoter, thereby decreasing the expression of ECRG4. Cardiomyocyte-specific loss of Ecrg4 significantly enriched the DEPs in the signaling pathways (oxidative phosphorylation, apoptosis, thermogenesis, and cardiac muscle contraction, among others) commonly involved in DOX-induced cardiotoxicity.

Studies on ECRG4 in atrial fibrillation have also been conducted in animal models. Huang et al. previously found that the expression of ECRG4 was significantly decreased in a rapid atrial pacing-induced canine AF model, suggesting that ECRG4 participates in the pathogenesis of AF (44). In addition, another study found that 24 hours after rapid electrical stimulation, ECRG4 was significantly downregulated in mouse atrial myocytes HL1 (50). Furthermore, The International Mouse Phenotyping Consortium revealed that ECRG4 knockout mice have a shorter QRS complex duration, highlighting the role of ECRG4 in heart rate/rhythm regulation. Collectively, the above findings indicate that both tachyarrhythmias and rapid pacing can induce a significant decrease in the expression of ECRG4, and the decrease in ECRG4 further leads to atrial remodeling, which is essential for the generation and maintenance of atrial fibrillation.

Atrial fibrillation and rhythm control regulated by ECRG4: Possible mechanisms

ECRG4 affects multiple cardiac ion channels expression

ECRG4 knockdown in neonatal atrial myocytes significantly upregulated the expression of calcium-binding protein family gene (s100a1, s100a8), while downregulating gap channel protein-1(Gjal) expression, leading to a significant shortening of action potential duration (APD50 and APD90) (Figure 2) (30). Calcium homeostasis plays an important regulatory role in myocardial remodeling; s100a1 and sl00a8, members of the S100 calcium-binding protein family and expressed by cardiomyocytes, are key to regulating the Ca2+ concentration in cardiomyocytes (51). and in patients with atrial fibrillation, the shortened duration of the action potential can trigger and result in a continuous reentry loop. The shortening of action potential duration is mainly caused by an increase in K+ outward current and/or a decrease in inward current induced by Ca2+ during repolarization, and in patients with atrial fibrillation, the shortened duration of the action potential can trigger and result in a continuous reentry loop (52). In human-induced pluripotent stem cell-derived cardiac cells (hiPSC-CMs), ECRG4 knockdown using siRNA also altered the expression of multiple ion channels: SCN5A (sodium channel), KCNH2 (HERG channel) and KCND3 (transient outward K channel) were reduced, while KCNN4 (SK4 channel) and HCN2 (funny channel) were increased (53). Taken together, the above studies suggest that ECRG4 may be involved in atrial fibrillation via regulating these channels.

ECRG4 affects the expression of matrix metalloproteinases

Studies have shown that Matrix metalloproteinase play an important role in the development of hypertension and atrial fibrillation by affecting the degradation of the extracellular matrix (54). MMP3 and MMP9 were increased in patients with recurrent AF within one year after electrical cardioversion (55). Studies have shown that MMP-9 is significantly higher in obese patients with paroxysmal atrial fibrillation than in obese patients alone. With the increase of MMP-9 in obese patients exceeding 285ng/ml, the occurrence of AF can be predicted with a sensitivity of 74.5% and specificity of 94% (56). It has been shown that ECRG4 can regulate the expression of MMPs in various organs and tissues. In oral squamous cell carcinoma, ECRG4 down-regulated the expression of matrix metalloproteinases (MMP-9 and MMP-13) (57). In atrial myocytes, ECRG4 knockdown significantly upregulated the expression of matrix metalloproteinase3 (MMP3) (30), which may contribute to atrial fibrillation.

ECRG4 and immune inflammatory reactions

A substantial amount of evidence suggests that the onset and progression of AF are strongly linked to inflammation (5861). Epidemiological studies have shown that compared with subjects with normal sinus rhythm, c-reactive protein and inflammatory cytokines such as TNF-α, IL-1β, IL-8, IL-6 and monocyte chemoattractant protein-1 (MCP-1) were significantly upregulated in patients with atrial fibrillation (59). Inflammatory mediators can disrupt cellular calcium homeostasis, activate and promote fibrosis, inhibit gap junction protein (Gjal)-mediated cell-cell communication (GJIC), and induce cardiomyocyte necrosis and apoptosis (58, 62, 63). ECRG4 knockdown in atrial myocytes significantly increased the production of pro-inflammatory genes. In recent years, a large amount of literature supports the involvement of ECRG4 in inflammation, injury, and infection. ECRG4 is a candidate chemokine that is highly expressed on leukocytes and regulates early neutrophil recruitment and subsequent CD44-mediated inflammatory decline, making ECRG4 a therapeutic target for inflammatory diseases (64). ECRG4 is also expressed on the cell surface of epithelial cells (12, 13, 65, 66). Cell surface ECRG4 is expressed in quiescent tissue and may have a “sentinel” function to monitor homeostasis, measure pro-inflammatory responses to injury and infection, and thus remain quiescent (1113, 65). When infection or inflammation occurs, the protease activates and initiates the downregulation of ECRG4 gene expression, which is released from the cell surface in a processed form (9, 11, 67). Other studies have shown that ECRG4 has a physiological role in measuring parenchymal and inflammatory responses to traumatic brain injury, with small needle wounds leading to a temporary reduction and full recovery of ECRG4 within 24-48 hours (13, 65). These findings suggest that low expression of ECRG4 may contribute to the development of atrial fibrillation through an inflammatory response.

ECRG4: Potential clinical target in tumor and arrhythmia

ECRG4 not only has a tumor suppressor effect but also cytokine-like functions. ECGG4 is found in a variety of bodily fluids, including blood, urine, saliva, pleural effusion, and cerebrospinal fluid. Decreased concentrations of ECRG4 in body fluids may indicate cancer development, suggesting that ECRG4 may be a biomarker for predicting cancer occurrence. Previous studies have found that methylated ECRG4 cDNA has promising diagnostic and clinical translational potential (68, 69). Meanwhile, promoter methylation determines ECRG4 expression status, and its aberration could help detect early cancer and predict severity (70). These findings show that ECRG4 can be used as a biomarker for cancer diagnosis as well as predicting staging and invasiveness. The wide distribution and diverse functions of ECRG4 make it an ideal target for drug therapy. ECRG4 has several functions, including tumor suppression, heart rhythm regulation, cardiac homeostasis maintenance, and involvement in the aging process. Overexpression of ECRG4 has been found to increase the sensitivity of gastric cancer cell line, SGC-7901 to 5-FU and NPC cell line, CNE1 to cisplatin, thereby improving the therapeutic effect of chemotherapy drugs (71, 72). Upregulation of ECRG4 expression or activity may also be used to treat diseases characterized by tissue dysfunction caused by attenuated ECRG4 expression. Since down-regulation of ECRG4 is associated with atrial fibrillation, up-regulating ECRG4 expression may aid in treating atrial fibrillation. Moreover, inhibition of ECRG4 can counteract senescence-associated cellular senescence. Interestingly, ECRG4 expression is generally silenced by promoter methylation, which demethylating agents can reactivate. There are currently two types of demethylation drugs, nucleoside DNMT inhibitors and non-nucleoside DNMT inhibitors, the efficacy of which is still being investigated. Furthermore, ECRG4 is a secretory protein that attaches to the cell surface and undergoes proteolysis to achieve biological activation. Therefore, high-affinity receptor agonists or protease inhibitors of ECRG4 are attractive targets for future drug development.

Perspectives

ECRG4 was initially known for its antitumor function, but as research progressed, its role in various physiology and pathology was gradually revealed, as was its role in the heart. The literature on atrial fibrillation induced by down-regulation of ECRG4 was summarized, and it was found that down-regulation of ECRG4 could induce atrial fibrillation through affecting ion channels, MMPs expression, and activating inflammatory response. ECRG4 is involved in the mechanism of doxorubicin-induced myocardial injury, which suggests that ECRG4 has myocardial protective function. In conclusion, ECRG4 can regulate rhythm, maintain cardiac homeostasis and protect myocardium. This study comprehensively reviewed the biological background of ECRG4 gene and its expression in cardiovascular system, focusing on the possible mechanism of ECRG4’s involvement in the formation of atrial fibrillation, which provides a new idea for reducing perioperative atrial fibrillation and myocardial injury in patients with esophageal cancer, gastric cancer and other tumors.

The uniqueness of ECRG4 makes it a potential target for precision medicine. Current research is primarily based on in vitro experiments, such as studies in KO or transgenic mouse models, which are extremely useful in determining the role of ECRG4 in vivo. The transition from basic research to clinical application necessitates a lengthy process of clarification and validation. Future research will need to decipher the mechanisms of action of ECRG4 and its signaling pathways. Continued development of new targeted drugs is expected to benefit not only the treatment of esophageal cancer, gastric cancer, and other cancers but also the treatment of cardiovascular diseases such as atrial fibrillation and myocardial injury.

Author contributions

ZZ, WW, and YZ contributed equally to this work. All authors contributed to the article and approved the submitted version.

Funding

This work was supported by the National Natural Science Foundation of China (82071233) and Shanghai Municipality in the 2021 “Science and Technology Innovation Action Plan” Sailing Plan (21YF1442900) and Nurture projects for basic research of Shanghai Chest Hospital (2019YNJCM08).

Acknowledgments

The authors wish to thank Ning Ma (DSc), Zhichao Liu (Ph D) from the Department of Esophageal Surgery and Zihan Li (Ph D) from the Department of Good Clinical Practice at Shanghai Chest Hospital, Shanghai, China, and also thank to the Figdraw drawing platform.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Zhou R, Liu Y, Huang W, Dang X. Potential functions of esophageal cancer-related gene-4 in the cardiovascular system. Front Med (2019) 13:639–45. doi: 10.1007/s11684-019-0701-0

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Li LW, Yu XY, Yang Y, Zhang CP, Guo LP, Lu SH. Expression of esophageal cancer related gene 4 (ECRG4), a novel tumor suppressor gene, in esophageal cancer and its inhibitory effect on the tumor growth in vitro and in vivo. Int J Cancer (2009) 125:1505–13. doi: 10.1002/ijc.24513

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Li LW, Li YY, Li XY, Zhang CP, Zhou Y, Lu SH. A novel tumor suppressor gene ECRG4 interacts directly with TMPRSS11A (ECRG1) to inhibit cancer cell growth in esophageal carcinoma. BMC Cancer (2011) 11:52. doi: 10.1186/1471-2407-11-52

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Mori Y, Ishiguro H, Kuwabara Y, Kimura M, Mitsui A, Kurehara H, et al. Expression of ECRG4 is an independent prognostic factor for poor survival in patients with esophageal squamous cell carcinoma. Oncol Rep (2007) 18:981–5. doi: 10.3892/or.18.4.981

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Erichsen R, Christiansen CF, Mehnert F, Weiss NS, Baron JA, Sørensen HT. Colorectal cancer and risk of atrial fibrillation and flutter: a population-based case-control study. Intern Emerg Med (2012) 7:431–8. doi: 10.1007/s11739-011-0701-9

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Zhang Z, Xu M, Wu D, Zhang X, Wu J. Postoperative myocardial injury in middle-aged and elderly patients following curative resection of esophageal cancer with aggressive or standard body temperature management: A randomized controlled trial. Anesth Analg (2019) 129:352–9. doi: 10.1213/ANE.0000000000004022

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Su T, Liu H, Lu S. [Cloning and identification of cDNA fragments related to human esophageal cancer]. Zhonghua Zhong Liu Za Zhi (1998) 20:254–7.

PubMed Abstract | Google Scholar

8. Mirabeau O, Perlas E, Severini C, Audero E, Gascuel O, Possenti R, et al. Identification of novel peptide hormones in the human proteome by hidden Markov model screening. Genome Res (2007) 17:320–7. doi: 10.1101/gr.5755407

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Dang X, Podvin S, Coimbra R, Eliceiri B, Baird A. Cell-specific processing and release of the hormone-like precursor and candidate tumor suppressor gene product, Ecrg4. Cell Tissue Res (2012) 348:505–14. doi: 10.1007/s00441-012-1396-6

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Wang LH, Wu CF, Rajasekaran N, Shin YK. Loss of tumor suppressor gene function in human cancer: An overview. Cell Physiol Biochem (2018) 51:2647–93. doi: 10.1159/000495956

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Baird A, Coimbra R, Dang X, Lopez N, Lee J, Krzyzaniak M, et al. Cell surface localization and release of the candidate tumor suppressor Ecrg4 from polymorphonuclear cells and monocytes activate macrophages. J Leukoc Biol (2012) 91:773–81. doi: 10.1189/jlb.1011503

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Kurabi A, Pak K, Dang X, Coimbra R, Eliceiri BP, Ryan AF, et al. Ecrg4 attenuates the inflammatory proliferative response of mucosal epithelial cells to infection. PloS One (2013) 8:e61394. doi: 10.1371/journal.pone.0061394

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Podvin S, Gonzalez AM, Miller MC, Dang X, Botfield H, Donahue JE, et al. Esophageal cancer related gene-4 is a choroid plexus-derived injury response gene: evidence for a biphasic response in early and late brain injury. PloS One (2011) 6:e24609. doi: 10.1371/journal.pone.0024609

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Chen L, Tang H, Liu G, Xiao S, Liang D, Ma J, et al. MicroRNA-196b promotes gastric cancer progression by targeting ECRG4. Anticancer Drugs (2021) 32:127–37. doi: 10.1097/CAD.0000000000000998

PubMed Abstract | CrossRef Full Text | Google Scholar

15. You Y, Hu S. Dysregulation of ECRG4 is associated with malignant properties and of prognostic importance in human gastric cancer. Cancer biomark (2022) 34:55–66. doi: 10.3233/CBM-210334

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Li C, Zhang P, Jiang A, Mao JH, Wei G. A short synthetic peptide fragment of human C2ORF40 has therapeutic potential in breast cancer. Oncotarget (2017) 8:41963–74. doi: 10.18632/oncotarget.16713

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Sabatier R, Finetti P, Adelaide J, Guille A, Borg JP, Chaffanet M, et al. Down-regulation of ECRG4, a candidate tumor suppressor gene, in human breast cancer. PloS One (2011) 6:e27656. doi: 10.1371/journal.pone.0027656

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Wu Y, Xiang Q, Lv X, Xiang X, Feng Z, Tian S, et al. C2orf40 inhibits hepatocellular carcinoma through interaction with UBR5. J Gastroenterol Hepatol (2021) 36:2581–91. doi: 10.1111/jgh.15441

PubMed Abstract | CrossRef Full Text | Google Scholar

19. You Y, Hu S. Aberrant expression of the esophageal carcinoma related gene 4 as a prognostic signature for hepatocellular carcinoma. Clin Res Hepatol Gastroenterol (2022) 46:101891. doi: 10.1016/j.clinre.2022.101891

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Xie Z, Li W, Ai J, Xie J, Zhang X. C2orf40 inhibits metastasis and regulates chemo-resistance and radio-resistance of nasopharyngeal carcinoma cells by influencing cell cycle and activating the PI3K/AKT/mTOR signaling pathway. J Transl Med (2022) 20:264. doi: 10.1186/s12967-022-03446-z

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Yang Z, Ye X, Zhang Y, Huang Y, Chen J, Zeng Y, et al. ECRG4 acts as a tumor suppressor in nasopharyngeal carcinoma by suppressing the AKT/GSK3β/β-catenin signaling pathway. Cytotechnology (2022) 74:231–43. doi: 10.1007/s10616-022-00520-8

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Zuo D, Liu G, Deng M, Gao Y. Efficacy of nedaplatin combined with docetaxel in patients with nasopharyngeal carcinoma and its influence on ECRG4 and VEGF expressions. J buon (2020) 25:1976–81.

PubMed Abstract | Google Scholar

23. Jia J, Dai S, Sun X, Sang Y, Xu Z, Zhang J, et al. A preliminary study of the effect of ECRG4 overexpression on the proliferation and apoptosis of human laryngeal cancer cells and the underlying mechanisms. Mol Med Rep (2015) 12:5058–64. doi: 10.3892/mmr.2015.4059

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Liang X, Gao J, Wang Q, Hou S, Wu C. ECRG4 represses cell proliferation and invasiveness via NFIC/OGN/NF-κB signaling pathway in bladder cancer. Front Genet (2020) 11:846. doi: 10.3389/fgene.2020.00846

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Rose M, Bringezu S, Godfrey L, Fiedler D, Gaisa NT, Koch M, et al. ITIH5 and ECRG4 DNA methylation biomarker test (EI-BLA) for urine-based non-invasive detection of bladder cancer. Int J Mol Sci (2020) 21:1117. doi: 10.3390/ijms21031117

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Götze S, Feldhaus V, Traska T, Wolter M, Reifenberger G, Tannapfel A, et al. ECRG4 is a candidate tumor suppressor gene frequently hypermethylated in colorectal carcinoma and glioma. BMC Cancer (2009) 9:447. doi: 10.1186/1471-2407-9-447

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Vanaja DK, Ballman KV, Morlan BW, Cheville JC, Neumann RM, Lieber MM, et al. PDLIM4 repression by hypermethylation as a potential biomarker for prostate cancer. Clin Cancer Res (2006) 12:1128–36. doi: 10.1158/1078-0432.CCR-05-2072

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Vanaja DK, Cheville JC, Iturria SJ, Young CY. Transcriptional silencing of zinc finger protein 185 identified by expression profiling is associated with prostate cancer progression. Cancer Res (2003) 63:3877–82.

PubMed Abstract | Google Scholar

29. Kujuro Y, Suzuki N, Kondo T. Esophageal cancer-related gene 4 is a secreted inducer of cell senescence expressed by aged CNS precursor cells. Proc Natl Acad Sci U.S.A. (2010) 107:8259–64. doi: 10.1073/pnas.0911446107

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Huang L, Yu H, Fan X, Li X, Mao L, Cheng J, et al. A potential role of esophageal cancer related gene-4 for atrial fibrillation. Sci Rep (2017) 7:2717. doi: 10.1038/s41598-017-02902-x

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Porzionato A, Rucinski M, Macchi V, Sarasin G, Malendowicz LK, De Caro R. ECRG4 expression in normal rat tissues: expression study and literature review. Eur J Histochem (2015) 59:2458. doi: 10.4081/ejh.2015.2458

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Allessie MA, Boyden PA, Camm AJ, Kléber AG, Lab MJ, Legato MJ, et al. Pathophysiology and prevention of atrial fibrillation. Circulation (2001) 103:769–77. doi: 10.1161/01.CIR.103.5.769

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Ostenfeld EB, Erichsen R, Pedersen L, Farkas DK, Weiss NS, Sørensen HT. Atrial fibrillation as a marker of occult cancer. PloS One (2014) 9:e102861. doi: 10.1371/journal.pone.0102861

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Conen D, Wong JA, Sandhu RK, Cook NR, Lee IM, Buring JE, et al. Risk of malignant cancer among women with new-onset atrial fibrillation. JAMA Cardiol (2016) 1:389–96. doi: 10.1001/jamacardio.2016.0280

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Botto F, Alonso-Coello P, Chan MT, Villar JC, Xavier D, Srinathan S, et al. Myocardial injury after noncardiac surgery: a large, international, prospective cohort study establishing diagnostic criteria, characteristics, predictors, and 30-day outcomes. Anesthesiology (2014) 120:564–78. doi: 10.1097/ALN.0000000000000113

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Tong C, Zheng J, Wu J. The effects of paravertebral blockade usage on pulmonary complications, atrial fibrillation and length of hospital stay following thoracoscopic lung cancer surgery. J Clin Anesth (2022) 79:110770. doi: 10.1016/j.jclinane.2022.110770

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Blencowe NS, Strong S, McNair AG, Brookes ST, Crosby T, Griffin SM, et al. Reporting of short-term clinical outcomes after esophagectomy: a systematic review. Ann Surg (2012) 255:658–66. doi: 10.1097/SLA.0b013e3182480a6a

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Low DE, Alderson D, Cecconello I, Chang AC, Darling GE, DʼJourno XB, et al. International consensus on standardization of data collection for complications associated with esophagectomy: Esophagectomy complications consensus group (ECCG). Ann Surg (2015) 262:286–94. doi: 10.1097/SLA.0000000000001098

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Mc Cormack O, Zaborowski A, King S, Healy L, Daly C, OʼFarrell N, et al. New-onset atrial fibrillation post-surgery for esophageal and junctional cancer: incidence, management, and impact on short- and long-term outcomes. Ann Surg (2014) 260:772–8. doi: 10.1097/SLA.0000000000000960

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Murthy SC, Law S, Whooley BP, Alexandrou A, Chu KM, Wong J. Atrial fibrillation after esophagectomy is a marker for postoperative morbidity and mortality. J Thorac Cardiovasc Surg (2003) 126:1162–7. doi: 10.1016/S0022-5223(03)00974-7

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Rao VP, Addae-Boateng E, Barua A, Martin-Ucar AE, Duffy JP. Age and neo-adjuvant chemotherapy increase the risk of atrial fibrillation following oesophagectomy. Eur J Cardiothorac Surg (2012) 42:438–43. doi: 10.1093/ejcts/ezs085

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Seesing MFJ, Scheijmans JCG, Borggreve AS, van Hillegersberg R, Ruurda JP. The predictive value of new-onset atrial fibrillation on postoperative morbidity after esophagectomy. Dis Esophagus (2018) 31. doi: 10.1093/dote/doy028

CrossRef Full Text | Google Scholar

43. Stawicki SP, Prosciak MP, Gerlach AT, Bloomston M, Davido HT, Lindsey DE, et al. Atrial fibrillation after esophagectomy: an indicator of postoperative morbidity. Gen Thorac Cardiovasc Surg (2011) 59:399–405. doi: 10.1007/s11748-010-0713-9

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Stippel DL, Taylan C, Schröder W, Beckurts KT, Hölscher AH. Supraventricular tachyarrhythmia as early indicator of a complicated course after esophagectomy. Dis Esophagus (2005) 18:267–73. doi: 10.1111/j.1442-2050.2005.00487.x

PubMed Abstract | CrossRef Full Text | Google Scholar

45. van der Sluis PC, Verhage RJ, van der Horst S, van der Wal WM, Ruurda JP, van Hillegersberg R. A new clinical scoring system to define pneumonia following esophagectomy for cancer. Dig Surg (2014) 31:108–16. doi: 10.1159/000357350

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Vaporciyan AA, Correa AM, Rice DC, Roth JA, Smythe WR, Swisher SG, et al. Risk factors associated with atrial fibrillation after noncardiac thoracic surgery: analysis of 2588 patients. J Thorac Cardiovasc Surg (2004) 127:779–86. doi: 10.1016/j.jtcvs.2003.07.011

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Weijs TJ, Seesing MF, van Rossum PS, Koëter M, van der Sluis PC, Luyer MD, et al. Internal and external validation of a multivariable model to define hospital-acquired pneumonia after esophagectomy. J Gastrointest Surg (2016) 20:680–7. doi: 10.1007/s11605-016-3083-5

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Kilickap S, Barista I, Akgul E, Aytemir K, Aksoy S, Tekuzman G. Early and late arrhythmogenic effects of doxorubicin. South Med J (2007) 100:262–5. doi: 10.1097/01.smj.0000257382.89910.fe

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Long D, Chen C, Li W, Peng W, Li D, Zhou R, et al. Cardiac expression of esophageal cancer-related gene-4 is regulated by Sp1 and is a potential early target of doxorubicin-induced cardiotoxicity. Cardiovasc Toxicol (2022) 22:404–18. doi: 10.1007/s12012-022-09722-0

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Perman JC, Boström P, Lindbom M, Lidberg U, StÅhlman M, Hägg D, et al. The VLDL receptor promotes lipotoxicity and increases mortality in mice following an acute myocardial infarction. J Clin Invest (2011) 121:2625–40. doi: 10.1172/JCI43068

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Kraus C, Rohde D, Weidenhammer C, Qiu G, Pleger ST, Voelkers M, et al. S100A1 in cardiovascular health and disease: closing the gap between basic science and clinical therapy. J Mol Cell Cardiol (2009) 47:445–55. doi: 10.1016/j.yjmcc.2009.06.003

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Iwasaki YK, Nishida K, Kato T, Nattel S. Atrial fibrillation pathophysiology: implications for management. Circulation (2011) 124:2264–74. doi: 10.1161/CIRCULATIONAHA.111.019893

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Lan H, Xu Q, El-Battrawy I, Li X, Zhao Z, Lang S, et al. Esophageal cancer related gene-4 affects multiple ion channel expression in human-induced stem cell-derived cardiomyocytes. Eur Heart J (2018) 39:813–. doi: 10.1093/eurheartj/ehy563.P3822

CrossRef Full Text | Google Scholar

54. Shin HJ, Park S, Yoon SJ, Choi DS, Cho DK, Kim JS, et al. Association between serum resistin and carotid intima media thickness in hypertension patients. Int J Cardiol (2008) 125:79–84. doi: 10.1016/j.ijcard.2007.02.028

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Mukherjee R, Akar JG, Wharton JM, Adams DK, McClure CD, Stroud RE, et al. Plasma profiles of matrix metalloproteinases and tissue inhibitors of the metalloproteinases predict recurrence of atrial fibrillation following cardioversion. J Cardiovasc Transl Res (2013) 6:528–35. doi: 10.1007/s12265-013-9471-2

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Podzolkov VI, Tarzimanova AI, Bragina AE, Gataulin RG, Oganesyan KA, Pokrovskaya AE, et al. [The importance of matrix metalloproteinases in the development of atrial fibrillation in obesity]. Ter Arkh (2021) 93:1451–6. doi: 10.26442/00403660.2021.12.201178

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Huang W, Zhou R, Mao L, Deng C, Dang X. Esophageal cancer related gene-4 inhibits the migration and proliferation of oral squamous cell carcinoma through BC200 lncRNA/MMP-9 and -13 signaling pathway. Cell Signal (2019) 62:109327. doi: 10.1016/j.cellsig.2019.05.012

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Harada M, Van Wagoner DR, Nattel S. Role of inflammation in atrial fibrillation pathophysiology and management. Circ J (2015) 79:495–502. doi: 10.1253/circj.CJ-15-0138

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Jiang Z, Dai L, Song Z, Li H, Shu M. Association between c-reactive protein and atrial fibrillation recurrence after catheter ablation: a meta-analysis. Clin Cardiol (2013) 36:548–54. doi: 10.1002/clc.22157

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Liu T, Li G, Li L, Korantzopoulos P. Association between c-reactive protein and recurrence of atrial fibrillation after successful electrical cardioversion: a meta-analysis. J Am Coll Cardiol (2007) 49:1642–8. doi: 10.1016/j.jacc.2006.12.042

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Tamariz L, Hernandez F, Bush A, Palacio A, Hare JM. Association between serum uric acid and atrial fibrillation: a systematic review and meta-analysis. Heart Rhythm (2014) 11:1102–8. doi: 10.1016/j.hrthm.2014.04.003

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Mesubi OO, Anderson ME. Atrial remodelling in atrial fibrillation: CaMKII as a nodal proarrhythmic signal. Cardiovasc Res (2016) 109:542–57. doi: 10.1093/cvr/cvw002

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Schotten U, Verheule S, Kirchhof P, Goette A. Pathophysiological mechanisms of atrial fibrillation: a translational appraisal. Physiol Rev (2011) 91:265–325. doi: 10.1152/physrev.00031.2009

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Dorschner RA, Lee J, Cohen O, Costantini T, Baird A, Eliceiri BP. ECRG4 regulates neutrophil recruitment and CD44 expression during the inflammatory response to injury. Sci Adv (2020) 6:eaay0518. doi: 10.1126/sciadv.aay0518

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Gonzalez AM, Podvin S, Lin SY, Miller MC, Botfield H, Leadbeater WE, et al. Ecrg4 expression and its product augurin in the choroid plexus: impact on fetal brain development, cerebrospinal fluid homeostasis and neuroprogenitor cell response to CNS injury. Fluids Barriers CNS (2011) 8:6. doi: 10.1186/2045-8118-8-6

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Shaterian A, Kao S, Chen L, DiPietro LA, Coimbra R, Eliceiri BP, et al. The candidate tumor suppressor gene Ecrg4 as a wound terminating factor in cutaneous injury. Arch Dermatol Res (2013) 305:141–9. doi: 10.1007/s00403-012-1276-7

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Ozawa A, Lick AN, Lindberg I. Processing of proaugurin is required to suppress proliferation of tumor cell lines. Mol Endocrinol (2011) 25:776–84. doi: 10.1210/me.2010-0389

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Giannopoulou L, Chebouti I, Pavlakis K, Kasimir-Bauer S, Lianidou ES. RASSF1A promoter methylation in high-grade serous ovarian cancer: A direct comparison study in primary tumors, adjacent morphologically tumor cell-free tissues and paired circulating tumor DNA. Oncotarget (2017) 8:21429–43. doi: 10.18632/oncotarget.15249

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Warton K, Mahon KL, Samimi G. Methylated circulating tumor DNA in blood: power in cancer prognosis and response. Endocr Relat Cancer (2016) 23:R157–71. doi: 10.1530/ERC-15-0369

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Wang YB, Ba CF. Promoter methylation of esophageal cancer-related gene 4 in gastric cancer tissue and its clinical significance. Hepatogastroenterology (2012) 59:1696–8. doi: 10.5754/hge12258

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Jiang CP, Wu BH, Wang BQ, Fu MY, Yang M, Zhou Y, et al. Overexpression of ECRG4 enhances chemosensitivity to 5-fluorouracil in the human gastric cancer SGC-7901 cell line. Tumour Biol (2013) 34:2269–73. doi: 10.1007/s13277-013-0768-1

PubMed Abstract | CrossRef Full Text | Google Scholar

72. You Y, Yang W, Qin X, Wang F, Li H, Lin C, et al. ECRG4 acts as a tumor suppressor and as a determinant of chemotherapy resistance in human nasopharyngeal carcinoma. Cell Oncol (Dordr) (2015) 38:205–14. doi: 10.1007/s13402-015-0223-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: esophageal cancer-related gene-4, myocardial injury, tumor suppressor gene, atrial fibrillation, radical surgery for esophageal cancer

Citation: Zhang Z, Wang W, Zhang Y, You X and Wu J (2023) A potential link between aberrant expression of ECRG4 and atrial fibrillation. Front. Oncol. 13:1031128. doi: 10.3389/fonc.2023.1031128

Received: 29 August 2022; Accepted: 08 February 2023;
Published: 22 February 2023.

Edited by:

Weian Zeng, Sun Yat-sen University Cancer Center (SYSUCC), China

Reviewed by:

Cheng Ni, Chinese Academy of Medical Sciences and Peking Union Medical College, China
Hui Zheng, Chinese Academy of Medical Sciences and Peking Union Medical College, China

Copyright © 2023 Zhang, Wang, Zhang, You and Wu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jingxiang Wu, wjx1132@163.com; Xingji You, yoyo1976@shu.edu.cn

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.