Open Access

Pathology and physiology of acid‑sensitive ion channels in the digestive system (Review)

  • Authors:
    • Li Zhang
    • Liming Zheng
    • Xingyue Yang
    • Shun Yao
    • Hui Wang
    • Jiaxing An
    • Hai Jin
    • Guorong Wen
    • Biguang Tuo
  • View Affiliations

  • Published online on: May 23, 2022     https://doi.org/10.3892/ijmm.2022.5150
  • Article Number: 94
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

As a major proton‑gated cation channel, acid‑sensitive ion channels (ASICs) can perceive large extracellular pH changes. ASICs play an important role in the occurrence and development of diseases of various organs and tissues including in the heart, brain, and gastrointestinal tract, as well as in tumor proliferation, invasion, and metastasis in acidosis and regulation of an acidic microenvironment. The permeability of ASICs to sodium and calcium ions is the basis of their physiological and pathological roles in the body. This review summarizes the physiological and pathological mechanisms of ASICs in digestive system diseases, which plays an important role in the early diagnosis, treatment, and prognosis of digestive system diseases related to ASIC expression.

1. Introduction

Maintaining normal proton concentrations inside and outside the cell is essential for the normal physiological functioning of numerous processes throughout the body (1). Acid-sensitive ion channels (ASICs) as effective proton sensors are sensitive to extracellular acidification (2). ASICs are widely expressed throughout the nervous system (2). Relatively more recently, they were discovered to be expressed in non-excitable tissues, such as in taste bud cells in the tongue contour nipple, human bone cells, vascular smooth muscle cells, lung epithelial cells, inner ear, and cochlear hair cells, among others (Fig. 1) (3). The function of ASICs in the nervous system has been widely studied (4), but there are considerably fewer studies assessing their roles in other systems of the body. Recent studies have found that ASICs are closely related to the physiological function and pathological development of diseases of the digestive system. In the digestive system, ASICs are involved in normal physiological functions such as gastrointestinal mechanical sensation and duodenal bicarbonate secretion (5,6). ASICs also underlie the development of gastrointestinal pains, gastro-intestinal cancer, and other pathological processes that involve a dysregulated acidic microenvironment or acidosis (7,8). This article reviews the research progress concerning ASICs in the digestive system.

2. Structure and function of ASICs

Molecular and structural characteristics of ASICs

In 1980, Krishtal and Pidoplichko (9) first recorded the cation currents triggered by a decrease in pH on the neuronal membrane. In 1997, Waldmann et al (10) first cloned H+-gated sensory neurons and named them ASICs. ASICs belong to the voltage-insensitive, amiloride-sensitive epithelial sodium channel (ENaC)/degenerin (DEG) channel family, which is a group of distantly related, non-voltage-gated Na+ channels found in animals. The epithelial channel ENaC in vertebrates has highly Na+-selectivity and low single-channel conductance, which can be blocked by the potassium retention diuretic, amiloride (11-14). At present, four subunits encoded by ASIC functional genes in humans and rodents have been identified; namely ASIC1a, ASIC1b, ASIC2a, ASIC2b, ASIC3, and ASIC4, where the a and b are subunits derived from the same respective gene but are different splice variants) (2,15,16). The spatial conformation of each subunit is similar to a 'clenched fist', with six domains: Wrist, finger, β-ball, thumb, joint, and a palm domain in the extracellular loop (15,17) (Fig. 2). The β-ball is the main region involved in sensing external protons and is termed the acidic pocket (2). Functional ASICs are aggregated by three homologous or heterologous subunits (18,19). Each ASIC subunit is composed of two hydrophobic transmembrane domains (TM1 and TM2), a large cysteine-rich extracellular loop, and a short conserved amino acid sequence at the C-terminal and N-terminal of the cell (10,17,20). Different subunits of ASICs have different properties, and the functional channels of the same trimer or heterotrimer are combined according to their own structural characteristics. The polymerization between different subunits allows for differing pH sensitivities, desensitization kinetics, and ion selectivity in the different ASICs (21). Due to these different electrophysiological properties, ASICs have a wide range of functions. Among the ASIC subunits, ASIC1a, ASIC1b, ASIC2a, and ASIC3 are directly controlled by protons, whereas ASIC2b and ASIC4 generally do not exhibit a functional role alone, and most ASIC2b and ASIC4 are auxiliary or regulatory subunits. ASIC2b and ASIC4 form oligomers with other subunits to allow them to exhibit their functional channel role (22). For example, ASIC2b mostly forms a heterotrimer with ASIC3 (23,24), and also forms a functional channel with ASIC1a during ischemic brain injury (25).

ASIC biological functions and features

Waldman and Lazdunski (12) recorded that when the extracellular pH dropped rapidly from 7.4 to <6.9, the proton concentration gradient on both sides of the channel changed, and the ASICs were activated. The activated ASICs showed permeability to cations and could allow the passage of sodium ions, calcium ions, and potassium ions (permeability index, sodium ions > calcium ions > potassium ions). ASICs generate characteristic instantaneous currents following proton-induced activation, and the current persistence is acid-dependent, thus the activated ASICs are rapidly deactivated (9,10,26). Therefore, ASICs, as a major proton-gated cation channel, are generally activated when the environmental pH drops. In recent years, significant progress has been made in the understanding of the structure and function of ASICs. ASICs have also been found to be activated by aprotic ligands (e.g., 2-guanidin-4-methylquinazoline) (27) at a normal physiological pH and this has been shown to be related to pain (21). Regarding the pH sensitivity of ASIC subunits, different subunits have different pH sensitivity ranges. Slight extracellular acidosis can activate ASIC1 and ASIC3 channels, whereas ASIC2a requires strong acidic conditions for activation. The pH-sensitive range of ASIC1a is 6.2-6.8, 5.1-6.2 for ASIC1b, 6.2-6.7 for ASIC3, and 4.1-5.0 for ASIC2a (28). Their pH sensitivity is related to His-72 and Gly-430 residues in the extracellular loop of ASIC protein (29). Therefore, in general, ASICs can perceive a large pH range. After activation of ASICs, Na+ and Ca2+ enters the cells, activating a series of pathophysiological processes (26). ASICs are widely expressed in various organs and tissues throughout the human body (Table I) (5,10,28,30-68). Thus far, ASICs have been found to be involved in various physiological processes such as in tactility (69), pain (3,70), olfaction (71), retinal integrity (48,72), learning and memory (73,74), fear (30,75), and anxiety (76). They are involved in the pathological process of various diseases in the human heart (77), brain (78), bone (79), gastrointestinal tract (80), and other organs and tissues.

Table I

Distribution of the expression of ASICs in tissues.

Table I

Distribution of the expression of ASICs in tissues.

ASICsExpressionPH0.5 activation(Refs.)
ASIC1aRat, mouse and human brain6.2-6.8(10,28,30,31)
Rat, mouse and human spinal cord(31-33)
Human lung epithelial cells(34)
Human bone cells(35)
Rat and mouse taste receptor cells(36)
Rat cultured vascular smooth muscle cells(37)
Human gliomas(38)
ASIC1bRat and mouse DRG5.1-6.2(28,31,39,40)
Rat taste receptor cells(41)
Rat carotid body(42)
Mouse cochlear hair cells(43)
ASIC2aRat, mouse and human brain4.1-5.0(28,31,44,45)
Rat, mouse and guinea pig DRG and NG(45-47)
Rat and mouse spinal cord(31-33)
Rat, mouse and rabbit retina(48-50)
Mice spiral ganglion in the cochlea(51)
Rat astrocytes(52)
Rat microglia(53)
Human bone cells(35-48)
Human lung epithelial cells(34)
Rat cultured vascular smooth muscle cells(37)
Rat carotid body(42)
Human gliomas(38)
Rat taste receptor cell(36,54)
ASIC2bRat and human brainNA(45,55)
Rat and mouse spinal cord(32,33)
Rat and mouse DRG(56,57)
Guinea pig NG and JG(58)
Rat and mouse retina(48,49)
Rat taste receptor cells(54)
ASIC3Rat, mouse and human DRG, TG and NG6.2-6.7(5,28,31,39)
Human brain, spinal cord, testis(55,59)
Rat and guinea pig vagal and glossopharyngeal ganglia(58)
Rat, mouse and rabbit retina(49,50,60)
Mouse chondrocytes and synoviocytes(61,62)
Rat brain(63)
Rat astrocytes(52)
Rat microglia(53)
Mouse adipocytes(64)
Human lung epithelial cells(34)
Human bone, cartilage and teeth(35,65)
Rat cultured vascular smooth muscle cells(35,37)
Rat and mouse taste receptor cells(36,61)
Mouse inner ear(66)
Rat carotid body(42)
ASIC4Mouse brain and spinal cordNA(31)
Human brain, inner ear and pituitary gland(67)
Mouse immune cells(68)
Rat and rabbit retina(49,50)

[i] ASICs, acid-sensitive ion channels; DRG, dorsal root ganglia; TG, trigeminal ganglia; JG, jugular ganglia; NG, nodular ganglia; NA, not available.

3. Related physiology of ASICs in the digestive system

ASICs and gastrointestinal acid-sensitive neurons

The stomach contains an abundance of glands, and the high concentration of HCl produced plays an important role in the normal digestive process (81); however, the strongly acidic environment is also a factor leading to certain gastrointestinal diseases (82). The erosion of the gastric mucosa by the excessively high concentration of gastric acid can cause a series of digestive symptoms (22). To enable gastric acid to play a physiological role without damaging the gastrointestinal mucosa, a cell capable of sensing acid is needed for fine regulation of the pH, in which epithelial cells and acid-sensitive neurons play a special role (22). Previous studies suggested that the vagus nerve pathway mainly regulates autonomic function, but recent studies have shown that the vagus nerve pathway may contribute to acid-sensing. Molecular and electrophysiological studies have shown that vagal sensory neurons projected to the proximal gastrointestinal tract express proton-gated ion channels (83,84). Krishtal and Pidoplichko (85) observed that sensory neurons could respond to protons nearby. Later, a large number of experimental studies confirmed this finding and indicated that acids could stimulate primary afferent neurons. Proton-gated ASICs are highly sensitive acid sensors. Although ASICs are rapidly deactivated after activation, they can still monitor long-term acidosis. This plays an important role in the gastrointestinal tract, which is rich in gastric acid. Whole-cell voltage-clamp recordings of dorsal root ganglia (DRG) and nodular ganglia (NG) neurons showed that proton-gated currents are related to ASICs (84). ASICs in the gastrointestinal tract primarily occur in the peripheral fibers of exogenous primary afferent neurons derived from the DRG and NG, and studies have shown that NG and DRG neurons in rats express ASIC3 (22,86). ASIC1, ASIC2, and ASIC3 are expressed in the DRG of thoracolumbar vertebrae that project into the colons of mice (87). When the ASIC3 gene was knocked out in mice, acid-induced excitability of gastric and esophageal vagus nerve fibers decreased (85). ASIC-mediated currents can be recorded from DRG neurons (88). ASICs are widely present in the digestive tract where they act as acid sensors and play an important role in acid monitoring of the gastrointestinal tract (2). During this process, ASICs perceive a decrease in extracellular PH, which stimulates acid-sensitive neurons and excites acid-sensitive neurons, thereby initiating the body's acid balance regulation system to maintain normal physiological functions (22).

ASICs and gastrointestinal mechanical sensation

Two major groups of mechanoreceptors in the vagus nerves of the stomach and esophagus of mice elicit signals of muscle stretching and mucosal touch (89), while two major groups in the innervation of the colon of mice elicit signals of serosal and mesenteric deformation (90). These mechanosensitive groups not only play a role in food intake and pain, but also in the regulation of digestive reflexes. On the basis of this experimental conclusion, Page et al (5) determined the expression of ASIC1a, 2, and 3 in the vagus nerve and DRG of mice using ASIC gene knockout mice, and proved that ASICs were involved in gastro-intestinal mechanoreception. Their further studies have shown that ASIC1a contributes to gastroesophageal and colonic afferent mechanotransduction, and increased sensitivity to all afferent mechanotransduction sites of visceral colonic afferent and vagus nerves after ASIC1a destruction. Conversely, in ASIC-deficient mice, except for gastroesophageal mucosal receptors, the mechanical sensitivity of all afferent classes was significantly reduced. For ASIC2, there was a notable difference in the different afferent subtypes. After ASIC2 was destroyed, the stomach showed an increase in the mechanical sensitivity of the gastroesophageal mucosal endings and a decrease in the gastroesophageal tension receptor, whereas, for the colon, it showed an increase in the colonic serous endings and no change in the colonic mesenteric endings (5). This indicates that ASIC subtypes have different effects on gastro-intestinal mechanical receptors. These ASIC gene knockout experiments affected mechanical conduction, and this resulted in a change in the gastrointestinal 'emptying mode' and affected food digestion.

The above studies show that ASICs are an important target for regulating gastrointestinal mechanical sensation. Several gastrointestinal diseases involving mechanical sensation can be treated by inhibiting or enhancing ASICs. For example, in irritable bowel syndrome, the stronger the expansion of the colon and rectum, the stronger the intestinal mechanical sensory signal, and the higher the perceived pain (91). If the expression of ASICs is weakened, the colorectal mechanical sensation signal is also weakened, and the pain will also be reduced. Similarly, in gastroesophageal reflux disease, if the expression of ASICs in the proximal stomach is lower, the proximal gastric mechanical sensory signal is reduced, the trigger of lower esophageal sphincter relaxation is weakened, and the reflux is also reduced (91).

ASICs and bicarbonate secretion in the duodenal mucosa

Through RT-PCR analysis, Dong et al (6) detected ASIC1a mRNA expression in the mouse duodenal epithelium and in the HT29 human intestinal epithelial cell line, and ASIC1a gene expression in mouse and human intestinal epithelial cells was confirmed. ASIC1a has notable permeability to extracellular calcium, and the lower the pH value of the solution is, the greater its permeability to calcium ions is. However, after the use of amiloride, an ASIC blocker, the increase in Ca2+ mobilization induced by acidosis was significantly weakened, indicating that the Ca2+ mobilization induced by acidosis was exerted via ASIC1a and ASIC1a played an important role in Ca2+ homeostasis in human intestinal epithelial cells. In their further research, they used acid as a physiological stimulus for duodenal lumen perfusion and compared amiloride to stimulation of the secretion of bicarbonate in the duodenal mucosa. According to the amount of bicarbonate secretion (DMBS) in the duodenal mucosa, it was concluded that acid may stimulate DMBS in mice in vivo by acting on duodenal ASIC1a. DMBS can maintain the weak alkaline environment of the digestive tract, provide a suitable pH environment for the activities of various digestive enzymes, neutralize gastric acid, and protect the intestinal mucosa. The experimental conclusion that acid promotes the secretion of bicarbonate in duodenal mucosa through ASICs may bring novel therapeutic targets for gastrointestinal diseases caused by increased acid.

For example, in duodenal ulcers, by enhancing the expression of ASICs, the secretion of duodenal bicarbonate also increases, which is beneficial to the improvement of symptoms in patients with duodenal ulcers.

4. Pathology of ASICs in the digestive system

ASICs and gastrointestinal pain

It has been well studied that central and peripheral nervous systems can regulate pain through ASICs (23,31,70,92,93). It has been found that ASICs are also associated with non-nerve associated pains. A decreased extracellular pH and inflammation are stimuli for pain. Local tissue acidification is also involved in the production of pain under pathological conditions. It may be that the acidic substances produced during inflammation reduce extracellular pH, activate proton-sensitive receptors/channels, regulate the function of pain-sensing neurons, and induce inflammatory pain allergy. Stimulating the gastric cavity of rats or mice with HCl concentrations exceeding physiological levels caused a visceral motor response, indicative of pain (94). Wultsch et al (95) eliminated the effects of ASIC3 gene disruption on the expression of c-Fos after gastric acid-induced neuronal excitability under gastric inflammation, whereas ASIC2 gene knockout did not alter the inflammatory hyperresponsiveness. Therefore, ASIC3 plays a role in the inflammatory hyperresponsiveness of gastric acid, which may also be related to ASIC3 leading to gastroesophageal reflux disease related to gastric acid reflux. Several studies have shown that primary neurons that dominate the small intestine and large intestine express acidic sensors such as ASICs (47,96). Previous experiments have shown that ASIC3 and TRPV1 contribute to the common functional visceral hypersensitivity in irritable bowel syndrome (7). After intracolonic injection of butyric acid (97), the colon of rats expanded without any inflammatory characteristics. Injection of the ASIC1a specific antagonist PcTx1 completely prevented the occurrence of colonic hypersensitivity (98), indicating that ASIC1 and ASIC3 were associated with the resultant colonic hypersensitivity. The more sensitive the colon expansion, the stronger the pain. A related study also confirmed that the high reactivity of colon dilatation was related to the upregulation of ASIC1a in colon DRG neurons and in the spinal cord (96,98). This established a novel direction for the treatment of patients with irritable bowel disease; reducing pain by weakening the expression of ASICs. Together, these studies suggest that ASICs play an important role in colonic hypersensitivity and hyperalgesia in irritable bowel patients.

ASICs and gastroesophageal reflux disease

As the symptoms of gastroesophageal reflux disease are closely related to acid reflux, the role of ASICs in the esophagus has attracted considerable attention. Studies have shown that ASIC1, ASIC2, and ASIC3 are expressed in the esophagus and its innervated nerves (5). The expression and function of each subtype of ASIC differ in the esophagus. ASIC1 mainly mediates the inhibitory response to acid and mechanical stimulation in the esophagus, whereas ASIC3 is expressed in esophageal spine cells and the mucosal muscle layer mediates high sensitivity to acid and inflammatory responses following acid stimulation and stimulation by strong mechanical expansion (99). Inflammatory mediators in the esophageal mucosa of gastroesophageal reflux disease patients can reduce the signal transduction threshold of ASICs, thereby mediating peripheral sensitization (100). Previous studies have shown that ASIC3 is closely related to the perception of pain, and more recent studies have found that they are also related to pain sensitivity. ASIC3 persistent expression in DRG neurons is involved in the formation of pain. With a decrease in the pain threshold and the emergence of pain sensitivity, ASIC3 expression is significantly upregulated (101,102). Inhibitors of ASICs can inhibit the pain allergy caused by peripheral acidification in rats and the pain caused by acidification in the human body (29), which further indicates the role of ASICs in pain sensitization. Although the pathogenesis of gastroesophageal reflux disease is complex, through in-depth studies of gastroesophageal reflux disease, it has been shown that visceral hypersensitivity and inflammation play an important role in the pathogenesis of gastroesophageal reflux disease (103). Recent studies have demonstrated that the upregulation of ASIC1 and ASIC3 expression is a possible cause of esophageal hypersensitivity to gastroesophageal reflux disease, providing a potential therapeutic target for patients with gastroesophageal reflux disease who do not respond to proton pump inhibitors (104). A gastroesophageal reflux disease rat model study found that during inflammation, the expression of ASIC1 and ASIC3 subunits was increased in the rat DRG (5). The above experiments show that ASICs are involved in gastroesophageal reflux disease caused by visceral hypersensitivity and inflammation. This discovery has provided a novel direction for the development of ASIC-related therapeutics for the management of gastroesophageal reflux disease.

ASICs and gastric cancer

Tumor tissues generally grow faster and metabolize vigorously, so local hypoxia and acidification of solid tumors are common phenomena and pathological features. An acidic microenvironment is an intrinsic feature of a tumor, and it promotes tumor invasion and metastasis (105). Several cancers are associated with the expression of ASICs, such as liver cancer, breast cancer, and glioma; ASIC expression in liver cancer is associated with its clinical stage (106,107). This suggests that ASICs are involved in the process of various tumor diseases. After activation of ASICs, extracellular signals can be introduced into cells, thereby regulating the expression of specific proteins in tumor cells. As mentioned above, there is an abundance of ASICs in the gastrointestinal tract, thus it is reasonable to speculate that the occurrence and development of gastric cancer may be related to ASICs. Chen et al (8) found that the expression of ASIC1a at the protein and mRNA level in gastric cancer tissues was higher than that in normal tissues, and that the upregulation of ASIC1a expression was positively correlated with advanced gastric cancer metastasis. ASIC silencing significantly inhibited the proliferation, migration, and invasion of gastric cancer cells in vitro, and the inhibition was affected by the acidity in the cell microenvironment, suggesting that a change in acidity could alter the tumorigenicity of cancer cells in vivo. This study also showed that ASIC1a was involved in the occurrence of gastric cancer, and that ASIC1a expression could affect the tumorigenicity of gastric cancer cells. Knockdown of ASIC1a in gastric cancer cells reduced the invasion and metastasis of gastric cancer. ASICs have been reported to activate autophagy (108,109). Zhang et al (110) found that ASIC1 and autophagy-associated protein 5 (ATG5) were expressed in the gastric cancer cell line SGC-7901. Downregulation of ATG5 or ASIC1 was able to inhibit the growth of gastric cancer cells, and ASIC1 also upregulated autophagy by activating ATG5 in gastric cancer cells. At the same time, it was found in the ASIC1-knockout mouse model that the downregulation of ASIC1 slowed down the growth of tumors and increased the survival time of mice. Although the current research on ASICs and gastric cancer is still in its infancy, these data show that inhibition of ASIC1 expression or inhibition of autophagy signaling pathways may serve as a novel means of targeted therapy for gastric cancer. At the same time, it may also play such a role in other types of cancers, thus highlighting novel avenues for the treatment of other types of cancer as well.

ASICs, liver fibrosis, and hepatolenticular degeneration

When inflammation is stimulated, oxidative stress and lipid peroxidation damage occur in the liver, leading to an imbalance in extracellular matrix (ECM) synthesis and degradation, and excessive ECM deposition in the liver tissue resulting in the formation of fibrosis (111).

The formation of hepatic fibrosis is related to the activation of hepatic stellate cells (112). ASIC1a is expressed in rat hepatic stellate cells, suggesting that the formation of liver fibrosis is related to ASIC1a (112). Zhu et al (113) found that ASIC1a expression was significantly increased in liver fibrosis, and miR-350 was involved in the formation of liver fibrosis regulated by ASIC1a and activation of stellate cells. Further analysis proved that the mechanism of hepatic fibrosis caused by ASIC1a was via ASIC1a-mediated regulation of m6A to affect the processing and modification of miR-350, and participate in hepatic fibrosis through modulation of the PI3K/AKT and ERK pathways (Fig. 3). This provided powerful insights into the potential treatment of liver fibrosis, highlighting the potential value of antagonists of PI3K/AKT and ERK signaling pathways to prevent and treat liver fibrosis. The early stages of hepatolenticular degeneration are the same as those for hepatic fibrosis, which is characterized by fibrosis of stellate cells. It was hypothesized that hepatolenticular degeneration may also be related to ASIC1a. Hepatolenticular degeneration is a disorder of copper metabolism. The disease is caused by a gene mutation of copper to Golgi body in the transfer cells, which leads to a disorder of copper metabolism. Excessive copper accumulates in the liver and brain, causing the corresponding systemic abnormalities (114). A large amount of copper accumulates in liver cells and this is toxic, leading to liver cell necrosis, inflammation, and activation of hepatic stellate cells to secrete ECM. Therefore, the early occurrence of hepatolenticular degeneration is the same as that of liver fibrosis. Kong et al (115) induced HSC-T6 activation with CuSO4, and found that ASIC1a was highly expressed in activated HSC-T6 cells. They speculated that ASIC1a played a role in promoting the process of hepatolenticular degeneration fibrosis. They also found high expression levels of ASIC1a in liver tissues of copper-loaded rats in animal experiments. Further analysis showed that ASIC1a in CuSO4-induced HSC-T6 cells may have regulated endoplasmic reticulum stress (ERS) via the P38MAPK pathway, thereby affecting hepatolenticular degeneration fibrosis and copper transport. Inhibition of ASIC1a-mediated ERS can improve copper transport, and the accumulation of copper in the liver and the degree of fibrosis in rats can be alleviated, which has a protective effect on Wilson's disease fibrosis. In summary, the above studies showed that liver fibrosis and hepatolenticular degeneration fibrosis are related to the activation of ASIC1a in an acidic environment, and the inhibition of ASIC1a may serve as a means of treatment of liver fibrosis.

ASICs and liver cancer

Hepatocellular carcinoma (HCC) mortality ranks third in global cancer mortality rates (116). β-catenin expression is common in cancer cells due to its ability to bind to intracellular surface cadherins to regulate cell adhesion. In cancer cells, the β-catenin/lymphoid enhancer factor (LEF)/T cell enhancer factor (TCF) axis is readily activated, leading to cancer cell proliferation (107). Jin et al (107) showed that ASIC1a could promote the excessive proliferation of liver cancer in vitro and in vivo through β-catenin activation and nuclear accumulation in an acidic environment. Knockdown/knockout of ASIC1a can inhibit the growth of liver cancer cells in vivo and in vitro, induce apoptosis of liver cancer cells, and induce cell cycle arrest of liver cancer by inhibiting LEF-TCF activity. This suggested that the mechanism by which ASIC1a resulted in the pathogenesis of liver cancer involved the activation of the β-catenin/LEF-TCF pathway to induce excessive proliferation of liver cancer cells. This finding provides a potential druggable target for the treatment of liver cancer. Quantitative studies have shown that an acidic microenvironment promotes cancer cell proliferation and migration (117). Jin et al (106) showed that the expression of ASIC1a in tumor tissues was significantly higher than that in non-tumor tissues based on immunohistochemical analysis, and that the expression in liver cancer tissues that exhibited postoperative metastasis was higher than that in liver cancer tissues without metastasis. Transwell assays showed that the mRNA and protein expression levels of ASIC1a in SMMC-7721 cells were significantly higher than those at pH 7.4 and 6.0 in a moderately acidic environment at pH 6.5, while the migration and invasion of SMMC-7721 cells were significantly inhibited after knockdown of ASIC1a. Additionally, in the same study, in 90 patients with liver cancer, it was found that ASIC1a expression in these patients was upregulated and was significantly correlated with a later clinical stage and a poorer prognosis. The above studies show that ASIC1a is significantly correlated with the migration and invasion of HCC, as well as a later clinical stage and a poorer prognosis. The higher the expression of ASIC1a in a moderately acidic environment, the more obvious the liver cancer symptoms were. Therefore, ASIC1a may serve as a marker for the diagnosis and prognosis of HCC. The inhibition of ASIC1a expression may highlight a novel direction for the treatment of liver cancer.

ASICs and pancreatic cancer

Pancreatic cancer is a common malignant epithelial tumor of the digestive system, with strong invasive properties. Epithelial-mesenchymal transition (EMT) plays an important role in tumor invasion and metastasis. In recent years, several studies have shown that the occurrence, development, invasion, and metastasis of pancreatic cancer show active EMT (118-120), but the specific mechanism of EMT in pancreatic cancer has not been elucidated. It has been previously reported that melanoma cells in acidic environments exhibit significant EMT-like characteristics in vitro and in vivo (121).

An acidic microenvironment induces EMT in pancreatic cancer cells by regulating the miR-652/ZEB1 pathway (122). An acidic microenvironment is also associated with ASICs, thus the relationship between ASICs and pancreatic cancer has attracted attention. Zhu et al (123) confirmed that ASIC1 and ASIC3 proteins are expressed in pancreatic cancer cell lines where they have a regulatory effect on pancreatic cancer cells in an acidic environment. In further experiments, they knocked out both ASIC1/ASIC3 and used amiloride to inhibit ASICs to reverse EMT of pancreatic cancer cells in an acidic environment, indicating that ASICs were involved in the process of EMT-induced pancreatic cancer. Calcium, as a second messenger in cells, is widely involved in various signaling pathways and plays an important role in regulating the invasion and migration of tumor cells (124). It was confirmed that ASIC1 and ASIC3 regulate EMT induced by an acidic environment via an increase in the intracellular Ca2+ concentration. RhoA is a member of the Rho family of small GTPases and plays a key role in cell invasion and metastasis (125). Ca2+ can regulate cytoskeletal remodeling by activating RhoA, thus playing a role in cell migration (126,127). Based on this theory, they showed that RhoA was activated by an ASIC1/ASIC3-[Ca2+]i pathway in an acidic environment to promote EMT of pancreatic cancer cells. In addition, stable knockdown of ASIC1 and ASIC3 in a nude mouse subcutaneously implanted tumor model significantly inhibited the invasion and metastasis of pancreatic cancer. In conclusion, they elucidated the complete signaling pathway of ASIC-induced pancreatic cancer; that is, an acidic environment induces EMT of pancreatic cancer cells through an ASIC1/3-[Ca2+]i-RhoA signaling pathway, resulting in increased invasion and metastasis of pancreatic cancer cells. The inhibition of ASIC1/3 can delay the progression of pancreatic cancer. It can also be used to manage pancreatic cancer by decreasing the Ca2+ concentration or inhibiting the RhoA protein.

ASICs and colorectal cancer (CRC)

An acidic extracellular microenvironment, namely acidosis caused by the Warburg effect (aerobic glycolysis) and a poor vasculature, is a biochemical feature of cancer (128). Acidosis changes the transcriptome characteristics of tumor cells, resulting in tumor cells suitable for survival, growth, and even metastasis in acidic environments (129). An increasing number of studies have shown that acidosis affects cancer progression by promoting tumor cell migration, invasion, metastasis, and angiogenesis (130-132). Previous studies have shown that acidosis promotes the invasion of CRC cells (131). On this basis, it is speculated that CRC may be related to ASICs. Zhou et al (80) verified the conjecture that ASIC2 promotes the invasion of CRC cells during acidosis. In further experiments, it was found that the average weight of tumors expressing ASIC2 was significantly higher than that of tumors in which ASIC2 expression was knocked down, indicating that ASIC2 promoted the proliferation of CRC cells in vitro and in vivo. At the same time, ASIC2 was also found to promote liver metastasis of CRC cells in vivo. Calcineurin is a Ca2+-dependent serine/threonine phosphatase with the immune central function that promotes the development of intestinal tumors by regulating the function of mouse tumor stem cells (133). When activated by intracellular Ca2+, calcineurin dephosphorylates the activated T nuclear factor (NFAT) protein, leading to nuclear translocation of NFAT (133). The transcription factors of the NFAT family play a key role in T-cell activation (134). The expression of IL-6 in NFAT1-deficient mice was significantly decreased, resulting in the occurrence and development of CRC (135,136). The results showed that ASIC2 overexpression significantly increased NFAT1 nuclear translocation induced by acidosis, while ASIC2 gene knockout had the opposite effect. In addition, CsA (a calcineurin inhibitor) inhibited calcineurin/NFAT signaling in a dose-dependent manner, which significantly reduced the invasion of SW480 cells induced by ASIC2 acidosis, and NFAT1 knockdown also inhibited the invasion of CRC cells. These results suggest that NFAT1 plays an important role in the invasion, migration, and metastasis of CRC by regulating gene transcription. Based on the above experimental results, ASIC2 promotes the invasion of CRC cells by activating the calcineurin/NFAT1 signaling pathway under acidosis. Further studies also showed that ASIC2 expression is related to CRC recurrence, tumor staging, distant metastasis, a poorer prognosis, and NFAT1 expression. If ASIC2 expression is inhibited, CRC can be delayed to a certain extent, highlighting a novel direction for the treatment of CRC. In summary, ASICs are associated with the occurrence, development, invasion, and metastasis of several digestive system tumors in acidosis, and this may underlie treatments for the prevention and treatment of digestive system tumors, and also open up avenues for novel research directions for tumors of other systems.

5. Conclusions

ASICs are widely distributed throughout the body where they function as acid sensors, widely involved in a variety of pathophysiological processes that involve acidosis. This review provides a basic and systematic summary of the physiological and pathological roles of ASICs in the digestive system, and shows that different subtypes of ASICs participate in the occurrence, development, invasion, and metastasis of digestive diseases and tumors in the acidic microenvironments, which is of great significance for further exploring the physiological role of ASICs in the body and developing targeted ASIC drug therapies. However, the extensive physiological and pathological mechanisms of ASICs in the human body have not been elucidated and require further study. The physiological and pathological significance of ASICs in the digestive system should be further studied and their relevance as molecular markers for the diagnosis and treatment of human-related diseases should be assessed.

Availability of data and materials

Not applicable.

Authors' contributions

LZ and LZ made substantial contributions to the conception and design of the article. XY, SY, HW, JA, HJ, GW, and BT were involved in revising the manuscript critically for important intellectual content. Data authentication is not applicable. All authors read and approved the final manuscript for publication.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

The present study was supported by grants from the National Natural Science Foundation of China (grant nos. 81960507, 82073087 and 82160112), the Science and Technology Bureau fund of Zunyi City [grant no. ZUN SHI KE HE HZ ZI (2019)93-Hao] and the Science and Technology Plan Project of Guizhou Province [grant nos. QIAN KE HE JI CHU-ZK(2021)YI BAN451 and QIAN KE HE LH ZI(2017)7095 HAO].

References

1 

Levin LR and Buck J: Physiological roles of acid-base sensors. Annu Rev Physiol. 77:347–362. 2015. View Article : Google Scholar

2 

Cheng YR, Jiang BY and Chen CC: Acid-sensing ion channels: Dual function proteins for chemo-sensing and mechano-sensing. J Biomed Sci. 25:462018. View Article : Google Scholar : PubMed/NCBI

3 

Deval E and Lingueglia E: Acid-Sensing Ion Channels and nociception in the peripheral and central nervous systems. Neuropharmacology. 94:49–57. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Chu XP and Xiong ZG: Physiological and pathological functions of acid-sensing ion channels in the central nervous system. Curr Drug Targets. 13:263–271. 2012. View Article : Google Scholar :

5 

Page AJ, Brierley SM, Martin CM, Price MP, Symonds E, Butler R, Wemmie JA and Blackshaw LA: Different contributions of ASIC channels 1a, 2, and 3 in gastrointestinal mechanosensory function. Gut. 54:1408–1415. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Dong X, Ko KH, Chow J, Tuo B, Barrett KE and Dong H: Expression of acid-sensing ion channels in intestinal epithelial cells and their role in the regulation of duodenal mucosal bicarbonate secretion. Acta Physiol (Oxf). 201:97–107. 2011. View Article : Google Scholar

7 

Jones RC III, Otsuka E, Wagstrom E, Jensen CS, Price MP and Gebhart GF: Short-term sensitization of colon mechanoreceptors is associated with long-term hypersensitivity to colon distention in the mouse. Gastroenterology. 133:184–194. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Chen X, Sun X, Wang Z, Zhou X, Xu L, Li F, Zhang X, Pan J, Qi L, Qian H and Mao Z: Involvement of acid-sensing ion channel 1a in gastric carcinoma cell migration and invasion. Acta Biochim Biophys Sin (Shanghai). 50:440–446. 2018. View Article : Google Scholar

9 

Krishtal OA and Pidoplichko VI: A receptor for protons in the nerve cell membrane. Neuroscience. 5:2325–2327. 1980. View Article : Google Scholar : PubMed/NCBI

10 

Waldmann R, Champigny G, Bassilana F, Heurteaux C and Lazdunski M: A proton-gated cation channel involved in acid-sensing. Nature. 386:173–177. 1997. View Article : Google Scholar : PubMed/NCBI

11 

Garty H and Palmer LG: Epithelial sodium channels: Function, structure, and regulation. Physiol Rev. 77:359–396. 1997. View Article : Google Scholar : PubMed/NCBI

12 

Waldmann R and Lazdunski M: H (+)-gated cation channels: Neuronal acid sensors in the NaC/DEG family of ion channels. Curr Opin Neurobiol. 8:418–424. 1998. View Article : Google Scholar : PubMed/NCBI

13 

Kellenberger S and Schild L: Epithelial sodium channel/degenerin family of ion channels: A variety of functions for a shared structure. Physiol Rev. 2:735–767. 2002. View Article : Google Scholar

14 

Benos DJ and Stanton BA: Functional domains within the degenerin/epithelial sodium channel (Deg/ENaC) super-family of ion channels. J Physiol. 520(Pt 3): 631–644. 1999. View Article : Google Scholar

15 

Kellenberger S and Schild L: International union of basic and clinical pharmacology. XCI. structure, function, and pharmacology of acid-sensing ion channels and the epithelial Na+ channel. Pharmacol Rev. 67:1–35. 2015. View Article : Google Scholar

16 

Sherwood TW, Frey EN and Askwith CC: Structure and activity of the acid-sensing ion channels. Am J Physiol Cell Physiol. 303:C699–C710. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Grunder S and Chen X: Structure, function, and pharmacology of acid-sensing ion channels (ASICs): Focus on ASIC1a. Int J Physiol Pathophysiol Pharmacol. 2:73–94. 2010.PubMed/NCBI

18 

Jasti J, Furukawa H, Gonzales EB and Gouaux E: Structure of acid-sensing ion channel 1 at 1.9 A resolution and low pH. Nature. 449:316–323. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Gonzales EB, Kawate T and Gouaux E: Pore architecture and ion sites in acid-sensing ion channels and P2X receptors. Nature. 460:599–604. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Krishtal O: The ASICs: Signaling molecules? modulators? Trends Neurosci. 26:477–483. 2003. View Article : Google Scholar

21 

Wemmie JA, Taugher RJ and Kreple CJ: Acid-sensing ion channels in pain and disease. Nat Rev Neurosci. 14:461–471. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Holzer P: Acid-sensing ion channels in gastrointestinal function. Neuropharmacology. 94:72–79. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Wemmie JA, Price MP and Welsh MJ: Acid-sensing ion channels: Advances, questions and therapeutic opportunities. Trends Neurosci. 29:578–586. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Holzer P: Acid-sensitive ion channels and receptors. Handb Exp Pharmacol. 283–332. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Sherwood TW, Lee KG, Gormley MG and Askwith CC: Heteromeric acid-sensing ion channels (ASICs) composed of ASIC2b and ASIC1a display novel channel properties and contribute to acidosis-induced neuronal death. J Neurosci. 31:9723–9734. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Waldmann R, Bassilana F, de Weille J, Champigny G, Heurteaux C and Lazdunski M: Molecular cloning of a non-inactivating proton-gated Na+ channel specific for sensory neurons. J Biol Chem. 272:20975–20978. 1997. View Article : Google Scholar : PubMed/NCBI

27 

Yu Y, Chen Z, Li WG, Cao H, Feng EG, Yu F, Liu H, Jiang H and Xu TL: A nonproton ligand sensor in the acid-sensing ion channel. Neuron. 68:61–72. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Noel J, Salinas M, Baron A, Diochot S, Deval E and Lingueglia E: Current perspectives on acid-sensing ion channels: New advances and therapeutic implications. Expert Rev Clin Pharmacol. 3:331–346. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Diochot S, Salinas M, Baron A, Escoubas P and Lazdunski M: Peptides inhibitors of acid-sensing ion channels. Toxicon. 49:271–284. 2007. View Article : Google Scholar

30 

Wemmie JA, Askwith CC, Lamani E, Cassell MD, Freeman JH Jr and Welsh MJ: Acid-sensing ion channel 1 is localized in brain regions with high synaptic density and contributes to fear conditioning. J Neurosci. 23:5496–5502. 2003. View Article : Google Scholar : PubMed/NCBI

31 

Chen CC and Zimmer A, Sun WH, Hall J, Brownstein MJ and Zimmer A: A role for ASIC3 in the modulation of high-intensity pain stimuli. Proc Natl Acad Sci USA. 99:8992–8997. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Baron A, Voilley N, Lazdunski M and Lingueglia E: Acid sensing ion channels in dorsal spinal cord neurons. J Neurosci. 28:1498–1508. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Wu LJ, Duan B, Mei YD, Gao J, Chen JG, Zhuo M, Xu L, Wu M and Xu TL: Characterization of acid-sensing ion channels in dorsal horn neurons of rat spinal cord. J Biol Chem. 279:43716–43724. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Su X, Li Q, Shrestha K, Cormet-Boyaka E, Chen L, Smith PR, Sorscher EJ, Benos DJ, Matalon S and Ji HL: Interregulation of proton-gated Na(+) channel 3 and cystic fibrosis transmembrane conductance regulator. J Biol Chem. 281:36960–36968. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Jahr H, van Driel M, van Osch GJ, Weinans H and van Leeuwen JP: Identification of acid-sensing ion channels in bone. Biochem Biophys Res Commun. 337:349–354. 2005. View Article : Google Scholar : PubMed/NCBI

36 

Richter TA, Dvoryanchikov GA, Roper SD and Chaudhari N: Acid-sensing ion channel-2 is not necessary for sour taste in mice. J Neurosci. 24:4088–4091. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Grifoni SC, Jernigan NL, Hamilton G and Drummond HA: ASIC proteins regulate smooth muscle cell migration. Microvasc Res. 75:202–210. 2008. View Article : Google Scholar

38 

Berdiev BK, Xia J, McLean LA, Markert JM, Gillespie GY, Mapstone TB, Naren AP, Jovov B, Bubien JK, Ji HL, et al: Acid-sensing ion channels in malignant gliomas. J Biol Chem. 278:15023–15034. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Chen CC, England S, Akopian AN and Wood JN: A sensory neuron-specific, proton-gated ion channel. Proc Natl Acad Sci USA. 95:10240–10245. 1998. View Article : Google Scholar : PubMed/NCBI

40 

Page AJ, Brierley SM, Martin CM, Martinez-Salgado C, Wemmie JA, Brennan TJ, Symonds E, Omari T, Lewin GR, Welsh MJ and Blackshaw LA: The ion channel ASIC1 contributes to visceral but not cutaneous mechanoreceptor function. Gastroenterology. 127:1739–1747. 2004. View Article : Google Scholar : PubMed/NCBI

41 

Liu L and Simon SA: Acidic stimuli activates two distinct pathways in taste receptor cells from rat fungiform papillae. Brain Res. 923:58–70. 2001. View Article : Google Scholar : PubMed/NCBI

42 

Tan ZY, Lu Y, Whiteis CA, Benson CJ, Chapleau MW and Abboud FM: Acid-sensing ion channels contribute to transduction of extracellular acidosis in rat carotid body glomus cells. Circ Res. 101:1009–1019. 2007. View Article : Google Scholar : PubMed/NCBI

43 

Ugawa S, Inagaki A, Yamamura H, Ueda T, Ishida Y, Kajita K, Shimizu H and Shimada S: Acid-sensing ion channel-1b in the stereocilia of mammalian cochlear hair cells. Neuroreport. 17:1235–1239. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Garcia-Anoveros J, Derfler B, Neville-Golden J, Hyman BT and Corey DP: BNaC1 and BNaC2 constitute a new family of human neuronal sodium channels related to degenerins and epithelial sodium channels. Proc Natl Acad Sci USA. 94:1459–1464. 1997. View Article : Google Scholar : PubMed/NCBI

45 

Lingueglia E, de Weille JR, Bassilana F, Heurteaux C, Sakai H, Waldmann R and Lazdunski M: A modulatory subunit of acid sensing ion channels in brain and dorsal root ganglion cells. J Biol Chem. 272:29778–29783. 1997. View Article : Google Scholar : PubMed/NCBI

46 

Price MP, Lewin GR, McIlwrath SL, Cheng C, Xie J, Heppenstall PA, Stucky CL, Mannsfeldt AG, Brennan TJ, Drummond HA, et al: The mammalian sodium channel BNC1 is required for normal touch sensation. Nature. 407:1007–1011. 2000. View Article : Google Scholar : PubMed/NCBI

47 

Hughes PA, Brierley SM, Young RL and Blackshaw LA: Localization and comparative analysis of acid-sensing ion channel (ASIC1, 2, and 3) mRNA expression in mouse colonic sensory neurons within thoracolumbar dorsal root ganglia. J Comp Neurol. 500:863–875. 2007. View Article : Google Scholar

48 

Ettaiche M, Guy N, Hofman P, Lazdunski M and Waldmann R: Acid-sensing ion channel 2 is important for retinal function and protects against light-induced retinal degeneration. J Neurosci. 24:1005–1012. 2004. View Article : Google Scholar : PubMed/NCBI

49 

Lilley S, LeTissier P and Robbins J: The discovery and characterization of a proton-gated sodium current in rat retinal ganglion cells. J Neurosci. 24:1013–1022. 2004. View Article : Google Scholar : PubMed/NCBI

50 

Brockway LM, Zhou ZH, Bubien JK, Jovov B, Benos DJ and Keyser KT: Rabbit retinal neurons and glia express a variety of ENaC/DEG subunits. Am J Physiol Cell Physiol. 283:C126–C134. 2002. View Article : Google Scholar : PubMed/NCBI

51 

Peng BG, Ahmad S, Chen S, Chen P, Price MP and Lin X: Acid-sensing ion channel 2 contributes a major component to acid-evoked excitatory responses in spiral ganglion neurons and plays a role in noise susceptibility of mice. J Neurosci. 24:10167–10175. 2004. View Article : Google Scholar : PubMed/NCBI

52 

Huang C, Hu ZL, Wu WN, Yu DF, Xiong QJ, Song JR, Shu Q, Fu H, Wang F and Chen JG: Existence and distinction of acid-evoked currents in rat astrocytes. Glia. 58:1415–1424. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Yu XW, Hu ZL, Ni M, Fang P, Zhang PW, Shu Q, Fan H, Zhou HY, Ni L, Zhu LQ, et al: Acid-sensing ion channels promote the inflammation and migration of cultured rat microglia. Glia. 63:483–496. 2015. View Article : Google Scholar

54 

Ugawa S, Yamamoto T, Ueda T, Ishida Y, Inagaki A, Nishigaki M and Shimada S: Amiloride-Insensitive currents of the acid-sensing ion Channel-2a (ASIC2a)/ASIC2b heteromeric sour-taste receptor channel. J Neurosci. 23:3616–3622. 2003. View Article : Google Scholar : PubMed/NCBI

55 

Delaunay A, Gasull X, Salinas M, Noël J, Friend V, Lingueglia E and Deval E: Human ASIC3 channel dynamically adapts its activity to sense the extracellular pH in both acidic and alkaline directions. Proc Natl Acad Sci USA. 109:13124–13129. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Voilley N, de Weille J, Mamet J and Lazdunski M: Nonsteroid Anti-Inflammatory Drugs Inhibit Both the Activity and the Inflammation-Induced Expression of Acid-Sensing Ion Channels in Nociceptors. J Neurosci. 21:8026–8033. 2001. View Article : Google Scholar : PubMed/NCBI

57 

Price MP, McIlwrath SL, Xie J, Cheng C, Qiao J, Tarr DE, Sluka KA, Brennan TJ, Lewin GR and Welsh MJ: The DRASIC cation channel contributes to the detection of cutaneous touch and acid stimuli in mice. Neuron. 32:1071–1083. 2001. View Article : Google Scholar

58 

Dusenkova S, Ru F, Surdenikova L, Nassenstein C, Hatok J, Dusenka R, Banovcin P Jr, Kliment J, Tatar M and Kollarik M: The expression profile of acid-sensing ion channel (ASIC) subunits ASIC1a, ASIC1b, ASIC2a, ASIC2b, and ASIC3 in the esophageal vagal afferent nerve subtypes. Am J Physiol Gastrointest Liver Physiol. 307:G922–G930. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Babinski K, Le KT and Seguela P: Molecular cloning and regional distribution of a human proton receptor subunit with biphasic functional properties. J Neurochem. 72:51–57. 1999. View Article : Google Scholar : PubMed/NCBI

60 

Ettaiche M, Deval E, Pagnotta S, Lazdunski M and Lingueglia E: Acid-sensing ion channel 3 in retinal function and survival. Invest Ophthalmol Vis Sci. 50:2417–2426. 2009. View Article : Google Scholar : PubMed/NCBI

61 

Ikeuchi M, Kolker SJ, Burnes LA, Walder RY and Sluka KA: Role of ASIC3 in the primary and secondary hyperalgesia produced by joint inflammation in mice. Pain. 137:662–669. 2008. View Article : Google Scholar : PubMed/NCBI

62 

Kolker SJ, Walder RY, Usachev Y, Hillman J, Boyle DL, Firestein GS and Sluka KA: Acid-sensing ion channel 3 expressed in type B synoviocytes and chondrocytes modulates hyaluronan expression and release. Ann Rheum Dis. 69:903–909. 2010. View Article : Google Scholar

63 

Meng QY, Wang W, Chen XN, Xu TL and Zhou JN: Distribution of acid-sensing ion channel 3 in the rat hypothalamus. Neuroscience. 159:1126–1134. 2009. View Article : Google Scholar : PubMed/NCBI

64 

Huang SJ, Yang WS, Lin YW, Wang HC and Chen CC: Increase of insulin sensitivity and reversal of age-dependent glucose intolerance with inhibition of ASIC3. Biochem Biophys Res Commun. 371:729–734. 2008. View Article : Google Scholar : PubMed/NCBI

65 

Sole-Magdalena A, Revuelta EG, Menénez-Díaz I, Calavia MG, Cobo T, García-Suárez O, Pérez-Piñera P, De Carlos F, Cobo J and Vega JA: Human odontoblasts express transient receptor protein and acid-sensing ion channel mechanosensor proteins. Microsc Res Tech. 74:457–463. 2011. View Article : Google Scholar

66 

Hildebrand MS, de Silva MG, Klockars T, Rose E, Price M, Smith RJ, McGuirt WT, Christopoulos H, Petit C and Dahl HH: Characterisation of DRASIC in the mouse inner ear. Hear Res. 190:149–160. 2004. View Article : Google Scholar : PubMed/NCBI

67 

Grunder S, Geissler HS, Bassler EL and Ruppersberg JP: A new member of acid-sensing ion channels from pituitary gland. Neuroreport. 11:1607–1611. 2000. View Article : Google Scholar : PubMed/NCBI

68 

Friese MA, Craner MJ, Etzensperger R, Vergo S, Wemmie JA, Welsh MJ, Vincent A and Fugger L: Acid-sensing ion channel-1 contributes to axonal degeneration in autoimmune inflammation of the central nervous system. Nat Med. 13:1483–1489. 2007. View Article : Google Scholar : PubMed/NCBI

69 

Omerbasic D, Schuhmacher LN, Bernal Sierra YA, Smith ES and Lewin GR: ASICs and mammalian mechanoreceptor function. Neuropharmacology. 94:80–86. 2015. View Article : Google Scholar

70 

Deval E, Gasull X, Noël J, Salinas M, Baron A, Diochot S and Lingueglia E: Acid-sensing ion channels (ASICs): Pharmacology and implication in pain. Pharmacol Ther. 128:549–558. 2010. View Article : Google Scholar : PubMed/NCBI

71 

Vann KT and Xiong ZG: Acid-sensing ion channel 1 contributes to normal olfactory function. Behav Brain Res. 337:246–251. 2018. View Article : Google Scholar

72 

Ettaiche M, Deval E, Cougnon M, Lazdunski M and Voilley N: Silencing acid-sensing ion channel 1a alters cone-mediated retinal function. J Neurosci. 26:5800–5809. 2006. View Article : Google Scholar : PubMed/NCBI

73 

Wemmie JA, Chen J, Askwith CC, Hruska-Hageman AM, Price MP, Nolan BC, Yoder PG, Lamani E, Hoshi T, Freeman JH Jr and Welsh MJ: The acid-activated ion channel ASIC contributes to synaptic plasticity, learning, and memory. Neuron. 34:463–477. 2002. View Article : Google Scholar : PubMed/NCBI

74 

Yermolaieva O, Leonard AS, Schnizler MK, Abboud FM and Welsh MJ: Extracellular acidosis increases neuronal cell calcium by activating acid-sensing ion channel 1a. Proc Natl Acad Sci USA. 101:6752–6757. 2004. View Article : Google Scholar : PubMed/NCBI

75 

Wemmie JA, Coryell MW, Askwith CC, Lamani E, Leonard AS, Sigmund CD and Welsh MJ: Overexpression of acid-sensing ion channel 1a in transgenic mice increases acquired fear-related behavior. Proc Natl Acad Sci USA. 101:3621–3626. 2004. View Article : Google Scholar : PubMed/NCBI

76 

Dwyer JM, Rizzo SJ, Neal SJ, Lin Q, Jow F, Arias RL, Rosenzweig-Lipson S, Dunlop J and Beyer CE: Acid sensing ion channel (ASIC) inhibitors exhibit anxiolytic-like activity in preclinical pharmacological models. Psychopharmacology (Berl). 203:41–52. 2009. View Article : Google Scholar

77 

Gibbons DD, Kutschke WJ, Weiss RM and Benson CJ: Heart failure induces changes in acid-sensing ion channels in sensory neurons innervating skeletal muscle. J Physiol. 593:4575–4587. 2015. View Article : Google Scholar : PubMed/NCBI

78 

Storozhuk M, Cherninskyi A, Maximyuk O, Isaev D and Krishtal O: Acid-sensing ion channels: Focus on physiological and some pathological roles in the brain. Curr Neuropharmacol. 19:1570–1589. 2021. View Article : Google Scholar : PubMed/NCBI

79 

Lee CY, Huang TJ, Wu MH, Li YY and Lee KD: High expression of acid-sensing ion channel 2 (ASIC2) in bone cells in osteoporotic vertebral fractures. Biomed Res Int. 2019:47142792019. View Article : Google Scholar : PubMed/NCBI

80 

Zhou ZH, Song JW, Li W, Liu X, Cao L, Wan LM, Tan YX, Ji SP, Liang YM and Gong F: The acid-sensing ion channel, ASIC2, promotes invasion and metastasis of colorectal cancer under acidosis by activating the calcineurin/NFAT1 axis. J Exp Clin Cancer Res. 36:1302017. View Article : Google Scholar : PubMed/NCBI

81 

Holzer P: Acid sensing by visceral afferent neurones. Acta Physiol (Oxf). 201:63–75. 2011. View Article : Google Scholar

82 

Kang JY and Yap I: Acid and gastric ulcer pain. J Clin Gastroenterol. 13:514–516. 1991. View Article : Google Scholar : PubMed/NCBI

83 

Dang K, Bielfeldt K, Lamb K and Gebhart GF: Gastric ulcers evoke hyperexcitability and enhance P2X receptor function in rat gastric sensory neurons. J Neurophysiol. 93:3112–3119. 2005. View Article : Google Scholar : PubMed/NCBI

84 

Sugiura T, Dang K, Lamb K, Bielefeldt K and Gebhart GF: Acid-sensing properties in rat gastric sensory neurons from normal and ulcerated stomach. J Neurosci. 25:2617–2627. 2005. View Article : Google Scholar : PubMed/NCBI

85 

Krishtal OA and Pidoplichko VI: A receptor for protons in the membrane of sensory neurons may participate in nociception. Neuroscience. 6:2599–2601. 1981. View Article : Google Scholar : PubMed/NCBI

86 

Schicho R, Florian W, Liebmann I, Holzer P and Lippe IT: Increased expression of TRPV1 receptor in dorsal root ganglia by acid insult of the rat gastric mucosa. Eur J Neurosci. 19:1811–1818. 2004. View Article : Google Scholar : PubMed/NCBI

87 

Bielefeldt K and Davis BM: Differential effects of ASIC3 and TRPV1 deletion on gastroesophageal sensation in mice. Am J Physiol Gastrointest Liver Physiol. 294:G130–G138. 2008. View Article : Google Scholar

88 

Leffler A, Monter B and Koltzenburg M: The role of the capsaicin receptor TRPV1 and acid-sensing ion channels (ASICS) in proton sensitivity of subpopulations of primary nociceptive neurons in rats and mice. Neuroscience. 139:699–709. 2006. View Article : Google Scholar : PubMed/NCBI

89 

Page AJ, Martin CM and Blackshaw LA: Vagal mechanoreceptors and chemoreceptors in mouse stomach and esophagus. J Neurophysiol. 87:2095–2103. 2002. View Article : Google Scholar : PubMed/NCBI

90 

Brierley SM, Jones RC III, Gebhart GF and Blackshaw LA: Splanchnic and pelvic mechanosensory afferents signal different qualities of colonic stimuli in mice. Gastroenterology. 127:166–178. 2004. View Article : Google Scholar : PubMed/NCBI

91 

Ruan N, Tribble J, Peterson AM, Jiang Q, Wang JQ and Chu XP: Acid-sensing ion channels and mechanosensation. Int J Mol Sci. 22:48102021. View Article : Google Scholar : PubMed/NCBI

92 

Bohlen CJ, Chesler AT, Sharif-Naeini R, Medzihradszky KF, Zhou S, King D, Sánchez EE, Burlingame AL, Basbaum AI and Julius D: A heteromeric Texas coral snake toxin targets acid-sensing ion channels to produce pain. Nature. 479:410–414. 2011. View Article : Google Scholar : PubMed/NCBI

93 

Kang S, Jang JH, Price MP, Gautam M, Benson CJ, Gong H, Welsh MJ and Brennan TJ: Simultaneous disruption of mouse ASIC1a, ASIC2 and ASIC3 genes enhances cutaneous mechanosensitivity. PLoS One. 7:e352252012. View Article : Google Scholar : PubMed/NCBI

94 

Lamb K, Kang YM, Gebhart GF and Bielefeldt K: Gastric inflammation triggers hypersensitivity to acid in awake rats. Gastroenterology. 125:1410–1418. 2003. View Article : Google Scholar : PubMed/NCBI

95 

Wultsch T, Painsipp E, Shahbazian A, Mitrovic M, Edelsbrunner M, Lazdunski M, Waldmann R and Holzer P: Deletion of the acid-sensing ion channel ASIC3 prevents gastritis-induced acid hyperresponsiveness of the stomach-brainstem axis. Pain. 134:245–253. 2008. View Article : Google Scholar

96 

Matricon J, Muller E, Accarie A, Meleine M, Etienne M, Voilley N, Busserolles J, Eschalier A, Lazdunski M, Bourdu S, et al: Peripheral contribution of NGF and ASIC1a to colonic hypersensitivity in a rat model of irritable bowel syndrome. Neurogastroenterol Motil. 25:e740–e754. 2013. View Article : Google Scholar : PubMed/NCBI

97 

Bourdu S, Dapoigny M, Chapuy E, Artigue F, Vasson MP, Dechelotte P, Bommelaer G, Eschalier A and Ardid D: Rectal instillation of butyrate provides a novel clinically relevant model of noninflammatory colonic hypersensitivity in rats. Gastroenterology. 128:1996–2008. 2005. View Article : Google Scholar : PubMed/NCBI

98 

Matricon J, Gelot A, Etienne M, Lazdunski M, Muller E and Ardid D: Spinal cord plasticity and acid-sensing ion channels involvement in a rodent model of irritable bowel syndrome. Eur J Pain. 15:335–343. 2011. View Article : Google Scholar

99 

Miwa H, Kondo T, Oshima T, Fukui H, Tomita T and Watari J: Esophageal sensation and esophageal hypersensitivity-overview from bench to bedside. J Neurogastroenterol Motil. 16:353–362. 2010. View Article : Google Scholar : PubMed/NCBI

100 

Guarino MP, Cheng L, Ma J, Harnett K, Biancani P, Altomare A, Panzera F, Behar J and Cicala M: Increased TRPV1 gene expression in esophageal mucosa of patients with non-erosive and erosive reflux disease. Neurogastroenterol Motil. 22:746–751 e219. 2010. View Article : Google Scholar : PubMed/NCBI

101 

Omori M, Yokoyama M, Matsuoka Y, Kobayashi H, Mizobuchi S, Itano Y, Morita K and Ichikawa H: Effects of selective spinal nerve ligation on acetic acid-induced nociceptive responses and ASIC3 immunoreactivity in the rat dorsal root ganglion. Brain Res. 1219:26–31. 2008. View Article : Google Scholar : PubMed/NCBI

102 

Staniland AA and McMahon SB: Mice lacking acid-sensing ion channels (ASIC) 1 or 2, but not ASIC3, show increased pain behaviour in the formalin test. Eur J Pain. 13:554–563. 2009. View Article : Google Scholar

103 

Yang M, Li ZS, Chen DF, Zou DW, Xu XR, Fang DC, Xu GM, Stephens RL and Wang ZG: Quantitative assessment and characterization of visceral hyperalgesia evoked by esophageal balloon distention and acid perfusion in patients with functional heartburn, nonerosive reflux disease, and erosive esophagitis. Clin J Pain. 26:326–331. 2010. View Article : Google Scholar : PubMed/NCBI

104 

Han X, Zhang Y, Lee A, Li Z, Gao J, Wu X, Zhao J, Wang H, Chen D, Zou D and Owyang C: Upregulation of acid sensing ion channels is associated with esophageal hypersensitivity in GERD. FASEB J. 36:e220832022. View Article : Google Scholar

105 

Webb BA, Chimenti M, Jacobson MP and Barber DL: Dysregulated pH: A perfect storm for cancer progression. Nat Rev Cancer. 11:671–677. 2011. View Article : Google Scholar : PubMed/NCBI

106 

Jin C, Ye QH, Yuan FL, Gu YL, Li JP, Shi YH, Shen XM, Bo-Liu and Lin ZH: Involvement of acid-sensing ion channel 1alpha in hepatic carcinoma cell migration and invasion. Tumour Biol. 36:4309–4317. 2015. View Article : Google Scholar : PubMed/NCBI

107 

Jin C, Yuan FL, Gu YL, Li X, Liu MF, Shen XM, Liu B and Zhu MQ: Over-expression of ASIC1a promotes proliferation via activation of the β-catenin/LEF-TCF axis and is associated with disease outcome in liver cancer. Oncotarget. 8:25977–25988. 2017. View Article : Google Scholar

108 

Sun X, Cao YB, Hu LF, Yang YP, Li J, Wang F and Liu CF: ASICs mediate the modulatory effect by paeoniflorin on α-synuclein autophagic degradation. Brain Res. 1396:77–87. 2011. View Article : Google Scholar : PubMed/NCBI

109 

Zhou RP, Wu XS, Wang ZS, Xie YY, Ge JF and Chen FH: Novel insights into acid-sensing ion channels: Implications for degenerative diseases. Aging Dis. 7:491–501. 2015. View Article : Google Scholar

110 

Zhang Q, Wu S, Zhu J, Chai D and Gan H: Down-regulation of ASIC1 suppressed gastric cancer via inhibiting autophagy. Gene. 608:79–85. 2017. View Article : Google Scholar : PubMed/NCBI

111 

Lee UE and Friedman SL: Mechanisms of hepatic fibrogenesis. Best Pract Res Clin Gastroenterol. 25:195–206. 2011. View Article : Google Scholar : PubMed/NCBI

112 

Wu FR, Pan CX, Rong C, Xia Q, Yuan FL, Tang J, Wang XY, Wang N, Ni WL and Chen FH: Inhibition of acid-sensing ion channel 1a in hepatic stellate cells attenuates PDGF-induced activation of HSCs through MAPK pathway. Mol Cell Biochem. 395:199–209. 2014. View Article : Google Scholar

113 

Zhu Y, Pan X, Du N, Li K, Hu Y, Wang L, Zhang J, Liu Y, Zuo L, Meng X, et al: ASIC1a regulates miR-350/SPRY2 by N6 -methyladenosine to promote liver fibrosis. FASEB J. 34:14371–14388. 2020. View Article : Google Scholar : PubMed/NCBI

114 

de Bie P, van de Sluis B, Burstein E, van de Berghe PV, Muller P, Berger R, Gitlin JD, Wijmenga C and Klomp LW: Distinct Wilson's disease mutations in ATP7B are associated with enhanced binding to COMMD1 and reduced stability of ATP7B. Gastroenterology. 133:1316–1326. 2007. View Article : Google Scholar : PubMed/NCBI

115 

Kong L, Huang H, Luan S, Liu H, Ye M and Wu F: Inhibition of ASIC1a-Mediated ERS improves the activation of HSCs and copper transport under copper load. Front Pharmacol. 12:6532722021. View Article : Google Scholar : PubMed/NCBI

116 

Yu LX and Schwabe RF: The gut microbiome and liver cancer: Mechanisms and clinical translation. Nat Rev Gastroenterol Hepatol. 14:527–539. 2017. View Article : Google Scholar : PubMed/NCBI

117 

Andersen AP, Moreira JM and Pedersen SF: Interactions of ion transporters and channels with cancer cell metabolism and the tumour microenvironment. Philos Trans R Soc Lond B Biol Sci. 369:201300982014. View Article : Google Scholar : PubMed/NCBI

118 

Javle MM, Gibbs JF, Iwata KK, Pak Y, Rutledge P, Yu J, Black JD, Tan D and Khoury T: Epithelial-mesenchymal transition (EMT) and activated extracellular signal-regulated kinase (p-Erk) in surgically resected pancreatic cancer. Ann Surg Oncol. 14:3527–3533. 2007. View Article : Google Scholar : PubMed/NCBI

119 

von Burstin J, Eser S, Paul MC, Seidler B, Brandl M, Messer M, von Werder A, Schmidt A, Mages J, Pagel P, et al: E-cadherin regulates metastasis of pancreatic cancer in vivo and is suppressed by a SNAIL/HDAC1/HDAC2 repressor complex. Gastroenterology. 137:361–371. 371.e1–5. 2009. View Article : Google Scholar : PubMed/NCBI

120 

Deng S, Zhu S, Wang B, Li X, Liu Y, Qin Q, Gong Q, Niu Y, Xiang C, Chen J, et al: Chronic pancreatitis and pancreatic cancer demonstrate active epithelial-mesenchymal transition profile, regulated by miR-217-SIRT1 pathway. Cancer Lett. 355:184–191. 2014. View Article : Google Scholar : PubMed/NCBI

121 

Peppicelli S, Bianchini F, Torre E and Calorini L: Contribution of acidic melanoma cells undergoing epithelial-to-mesenchymal transition to aggressiveness of non-acidic melanoma cells. Clin Exp Metastasis. 31:423–433. 2014. View Article : Google Scholar : PubMed/NCBI

122 

Deng S, Li X, Niu Y, Zhu S, Jin Y, Deng S, Chen J, Liu Y, He C, Yin T, et al: MiR-652 inhibits acidic microenvironment-induced epithelial-mesenchymal transition of pancreatic cancer cells by targeting ZEB1. Oncotarget. 6:39661–39675. 2015. View Article : Google Scholar : PubMed/NCBI

123 

Zhu S, Zhou HY, Deng SC, Deng SJ, He C, Li X, Chen JY, Jin Y, Hu ZL, Wang F, et al: ASIC1 and ASIC3 contribute to acidity-induced EMT of pancreatic cancer through activating Ca2+/RhoA pathway. Cell Death Dis. 8:e28062017. View Article : Google Scholar

124 

Prevarskaya N, Skryma R and Shuba Y: Calcium in tumour metastasis: New roles for known actors. Nat Rev Cancer. 11:609–618. 2011. View Article : Google Scholar : PubMed/NCBI

125 

Jaffe AB and Hall A: Rho GTPases: Biochemistry and biology. Annu Rev Cell Dev Biol. 21:247–269. 2005. View Article : Google Scholar : PubMed/NCBI

126 

Gulhati P, Bowen KA, Liu J, Stevens PD, Rychahou PG, Chen M, Lee EY, Weiss HL, O'Connor KL, Gao T and Evers BM: mTORC1 and mTORC2 regulate EMT, motility, and metastasis of colorectal cancer via RhoA and Rac1 signaling pathways. Cancer Res. 71:3246–3256. 2011. View Article : Google Scholar : PubMed/NCBI

127 

Fernandez-Tenorio M, Porras-González C, Castellano A, Del Valle-Rodríguez A, López-Barneo J and Ureña J: Metabotropic regulation of RhoA/Rho-associated kinase by L-type Ca2+ channels: New mechanism for depolarization-evoked mammalian arterial contraction. Circ Res. 108:1348–1357. 2011. View Article : Google Scholar : PubMed/NCBI

128 

Fais S, De Milito A, You H and Qin W: Targeting vacuolar H+-ATPases as a new strategy against cancer. Cancer Res. 67:10627–10630. 2007. View Article : Google Scholar : PubMed/NCBI

129 

Chen JL, Lucas JE, Schroeder T, Mori S, Wu J, Nevins J, Dewhirst M, West M and Chi JT: The genomic analysis of lactic acidosis and acidosis response in human cancers. PLoS Genet. 4:e10002932008. View Article : Google Scholar : PubMed/NCBI

130 

Moellering RE, Black KC, Krishnamurty C, Baggett BK, Stafford P, Rain M, Gatenby RA and Gillies RJ: Acid treatment of melanoma cells selects for invasive phenotypes. Clin Exp Metastasis. 25:411–425. 2008. View Article : Google Scholar : PubMed/NCBI

131 

Estrella V, Chen T, Lloyd M, Wojtkowiak J, Cornnell HH, Ibrahim-Hashim A, Bailey K, Balagurunathan Y, Rothberg JM, Sloane BF, et al: Acidity generated by the tumor microenvironment drives local invasion. Cancer Res. 73:1524–1535. 2013. View Article : Google Scholar : PubMed/NCBI

132 

Fukumura D, Xu L, Chen Y, Gohongi T, Seed B and Jain RK: Hypoxia and acidosis independently up-regulate vascular endothelial growth factor transcription in brain tumors in vivo. Cancer Res. 61:6020–6024. 2001.PubMed/NCBI

133 

Peuker K, Muff S, Wang J, Künzel S, Bosse E, Zeissig Y, Luzzi G, Basic M, Strigli A, Ulbricht A, et al: Epithelial calcineurin controls microbiota-dependent intestinal tumor development. Nat Med. 22:506–515. 2016. View Article : Google Scholar : PubMed/NCBI

134 

Chuvpilo S, Jankevics E, Tyrsin D, Akimzhanov A, Moroz D, Jha MK, Schulze-Luehrmann J, Santner-Nanan B, Feoktistova E, König T, et al: Autoregulation of NFATc1/A expression facilitates effector T cells to escape from rapid apoptosis. Immunity. 16:881–895. 2002. View Article : Google Scholar : PubMed/NCBI

135 

Weigmann B, Lehr HA, Yancopoulos G, Valenzuela D, Murphy A, Stevens S, Schmidt J, Galle PR, Rose-John S and Neurath MF: The transcription factor NFATc2 controls IL-6-dependent T cell activation in experimental colitis. J Exp Med. 205:2099–2110. 2008. View Article : Google Scholar : PubMed/NCBI

136 

Gerlach K, Daniel C, Lehr HA, Nikolaev A, Gerlach T, Atreya R, Rose-John S, Neurath MF and Weigmann B: Transcription factor NFATc2 controls the emergence of colon cancer associated with IL-6-dependent colitis. Cancer Res. 72:4340–4350. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2022
Volume 50 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang L, Zheng L, Yang X, Yao S, Wang H, An J, Jin H, Wen G and Tuo B: Pathology and physiology of acid‑sensitive ion channels in the digestive system (Review). Int J Mol Med 50: 94, 2022
APA
Zhang, L., Zheng, L., Yang, X., Yao, S., Wang, H., An, J. ... Tuo, B. (2022). Pathology and physiology of acid‑sensitive ion channels in the digestive system (Review). International Journal of Molecular Medicine, 50, 94. https://doi.org/10.3892/ijmm.2022.5150
MLA
Zhang, L., Zheng, L., Yang, X., Yao, S., Wang, H., An, J., Jin, H., Wen, G., Tuo, B."Pathology and physiology of acid‑sensitive ion channels in the digestive system (Review)". International Journal of Molecular Medicine 50.1 (2022): 94.
Chicago
Zhang, L., Zheng, L., Yang, X., Yao, S., Wang, H., An, J., Jin, H., Wen, G., Tuo, B."Pathology and physiology of acid‑sensitive ion channels in the digestive system (Review)". International Journal of Molecular Medicine 50, no. 1 (2022): 94. https://doi.org/10.3892/ijmm.2022.5150