International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Ion channels are a major class of membrane proteins that are sensitive to a variety of stimuli, including changes in membrane potential, ligand binding, temperature fluctuations and mechanical forces (1). These stimuli trigger conformational changes in the protein, leading to the formation of gated ion-permeable pores in biological membranes, which facilitate the passive diffusion of ions along their respective electrochemical gradients both across the cell membrane and between intracellular compartments. Ion channels contribute to the regulation of various processes, such as the maintenance of cellular osmotic pressure and membrane potential, motility (through interactions with the cytoskeleton), invasion, signal transduction, transcriptional activity and cell cycle progression, all of which can lead to tumor progression and metastasis (2). Their central role in intracellular and intercellular signaling also facilitates the coupling of extracellular events to cellular responses (3).
Over the past decade, interest in the study of ion channels in digestive disorders has increased. The literature suggests that dysregulated ion channel activity plays a pathogenic role in various digestive diseases, including cancer. For instance, dysfunction of ion channels and transporter proteins can lead to gastrointestinal mucosal damage, such as disruption of the bicarbonate and mucosal layers (4-7), loss of epithelial cells (8,9), atrophy of glandular mucosa (10), loss of tight junction (TJ) proteins (11-13), imbalances in the gut flora (14) and changes in mucosal blood flow (15). These alterations contribute to gastrointestinal mucosal diseases, including gastritis, ulcers, inflammatory bowel disease and gastrointestinal cancers (16).
Ion channels and transporter proteins have been reported to be crucial for maintaining both the exocrine and endocrine functions of the pancreas. They facilitate enzymatic reactions necessary for the digestion of fats and proteins in the intestines, while also facilitating the secretion of bicarbonate-rich fluids (17,18). Pancreatic islet β-cells express a variety of ion channels located in lipid membranes or subcellular organelles, which are collectively involved in glucose-dependent insulin secretion. Consequently, dysfunction of these ion channels and transporter proteins can result in pancreatic injury, potentially leading to conditions such as pancreatitis, diabetes mellitus, pancreatic cystic fibrosis (CF) and an increased risk of pancreatic cancer (19,20).
In addition, the role of ion channels and transporter proteins in liver diseases has garnered considerable attention. For instance, dysfunction of various calcium channels can disrupt intraand extracellular calcium homeostasis, leading to endoplasmic reticulum stress, mitochondrial dysfunction and the inhibition of autophagy in hepatocytes, all of which contribute to the development of nonalcoholic fatty liver disease (NAFLD) (21). Ion channels intersect with multiple signaling pathways, promoting hepatocellular carcinoma (HCC) cell invasion and metastasis by modulating gene expression, regulating cell proliferation and triggering epithelial-mesenchymal transition (22). In addition, ion channels and transporter proteins also regulate the growth and invasion of esophageal cancer (EC) (23-25).
As mentioned above, ion channels and transporter proteins play critical roles in regulating pathophysiological processes in various organs of the digestive system. While numerous studies have extensively investigated the role of cation channels, particularly calcium channels (26-32), sodium channels (33-36) and potassium channels (37-39), anion channels have received comparatively less attention. Anion channels (40,41) have been identified as important molecules with aberrant expression, activity and localization in various pathological conditions, including cardiovascular diseases, neurological disorders, metabolic diseases and cancer.
As the primary anion in the human body, intracellular chloride concentrations are dynamically regulated (42) and play a crucial role in maintaining intracellular and osmotic homeostasis, primarily through transmembrane transport and ion channels (43). Chloride channels have been implicated in a variety of pathological diseases (44). These channels and transport proteins are located both in the plasma membrane and intracellular membranes, with functions ranging from ion homeostasis and cell volume regulation to transepithelial transport and electrical excitability regulation (45). Chloride ions are regulated by chloride channels for their intra- and extracellular distribution and transport. Chloride channels are currently classified into several groups (Fig. 1), including the chloride channel (ClC) family, CF transmembrane conductance regulator (CFTR), calcium-activated chloride channels (CaCCs), volume-regulated anion channels (VRACs), proton-activated chloride channels (PACs) (46) and ligand-gated anion channels (47). Chloride channels have undergone significant functional differentiation and diversification during evolution and are distributed from prokaryotes to higher vertebrates (Fig. 2). The ClC family is one of the evolutionarily oldest classes of chloride channels and is widespread in prokaryotes and eukaryotes (48). During evolution, the ClC family differentiated into voltage-gated chloride channels and chloride/proton exchangers. In vertebrates, the ClC family has further differentiated into several isoforms (e.g., ClC-1 to -7), which are involved in the regulation of cell membrane potential or chloride homeostasis of intracellular organelles (49). Among CaCCs, anoctamin (ANO)1 has been extensively studied. ANO1 [transmembrane protein (TMEM)16A] belongs to the anoctamin family and is found in both invertebrates and vertebrates (50). ANO1 may have initially evolved as a lipid scramblase, and subsequently gained calcium-activated chloride channel function. In mammals, ANO1 is expressed in a variety of tissues and is involved in secretion, smooth muscle contraction and signaling (51). CFTR belongs to the ATP-binding cassette (ABC) superfamily of transporter proteins and is a vertebrate-specific chloride channel (52). CFTR has evolved to acquire a unique regulatory domain (R-domain) that allows it to be activated by cAMP-dependent phosphorylation (53). PACs/TMEM206 belongs to a family of transmembrane proteins, TMEM, that are highly conserved in vertebrates. No direct homologs have been identified in invertebrates, but functionally similar channels may exist in some lower organisms. In mammals, PACs are expressed in a variety of tissues, particularly in the brain, lungs and kidneys, and may be associated with pathological processes related to acidosis. PACs may have evolved as an adaptive mechanism for cells to cope with acidic environments, and are involved in maintaining intracellular pH homeostasis (54). Ligand-gated anion channels belong to the Cys-loop ligand-gated ion channel superfamily and share a common ancestor with related channels in invertebrates, such as glutamate-gated chloride channels (55). In vertebrates, gamma-aminobutyric acid type A receptor (GABA-A) and glycine receptor (GlyR) mediate inhibitory neurotransmission (56) (Fig. 2). Of note, in a study investigating chloride channels in an in vivo porcine model of esophageal acid damage, ClC-2 was found to localize to the basal epithelium of the porcine esophageal mucosa and esophageal submucosal glands. Chloride intracellular channel protein 1 recruited phosphatidylinositol 4-phosphate 5-kinases to lipid membranes, which, in turn, regulate cell-matrix adhesion and membrane protrusion, contributing to tumor aggressiveness, metastasis and a poor prognosis (57).
To date, there has been no comprehensive review of the role of chloride ion channels in digestive system diseases. Therefore, the aim of this review was to compile and summarize the physiopathological roles of chloride channels and transporter proteins in digestive system diseases, with the goal of providing a solid theoretical foundation for future research and offering new perspectives for the treatment of these diseases.
The ClC channel exists as a dimer, with each monomer forming a separate ion-conducting pore, and each monomer contains 18 alpha helices, some of which form ion-conducting pores (49). The ClC family comprises ClC-0, ClC-1, ClC-2, ClC-Ka, ClC-Kb, ClC-3, ClC-4, ClC-5, ClC-6 and ClC-7. On the basis of their distribution, ClC-0, ClC-1, ClC-2, ClC-Ka and ClC-Kb are plasma membrane channels, whereas the others are intracellular membrane channels. Functionally, ClC-1, ClC-2, ClC-Ka and ClC-Kb are voltage-gated chloride channels, whereas the remaining members act as Cl−/H+ exchangers. The ClC family is involved in several key functions, including volume regulation, ion dynamic homeostasis, transepithelial transport and the regulation of electrical excitability. ClC-2, a voltage-gated chloride channel composed of 907 amino acids with a molecular weight of 99 kDa, features a two-pore homodimeric structure (58,59). Its activity is regulated by changes in the extracellular pH, displaying a biphasic response. ClC-2 is activated by moderate acidification but abruptly shuts down when the pH drops below ~7.
ClC-2 mRNA levels are significantly elevated in patients with nonalcoholic steatohepatitis and mice lacking ClC-2 exhibit a beneficial metabolic phenotype, suggesting that reducing or knocking down ClC-2 may be a potential strategy for treating NAFLD and further research revealed that ClC-2 downregulated the activation of IRS-1/Akt/mTOR signaling (60) (Fig. 3).
Chloride channels also play a critical role in intestinal diseases. The intestinal barrier, which is essential for nutrient absorption and preventing the entry of harmful substances, consists of epithelial cells connected by apical junction complexes, comprising TJs and adherens junctions (AJs) (61,62). In a ClC-2 knockout mouse model, the absence of ClC-2 delayed recovery of intestinal barrier function after ischemic injury, possibly because ClC-2 anchors the TJ structure following injury (63). Additionally, disruption of AJs affected colon homeostasis and differentiation, promoting tumorigenicity through the regulation of β-catenin signaling (64). Furthermore, genetic inactivation variants in the ClC-Ka and ClC-Kb genes have been associated with renal salt-losing nephropathies, with or without deafness, including the rare Bartter's syndrome (BS) (65), highlighting the crucial role of the ClC-Ka and ClC-Kb genes in renal and inner ear chloride processing (66); however, to the best of our knowledge, no studies to date have explored the role of these chloride channels in digestive diseases.
ClC-3 plays an important role in cancer regulation as a volume-regulated anion channel that also controls the cell cycle and is overexpressed in various cancers. ClC-3 can promote the development and metastasis of colorectal cancer (CRC) through the Wnt/β-catenin signaling pathway (67) (Fig. 3). In a study on prognostic biomarkers in gastric cancer (GC), high expression of ClC-3 was associated with deeper tumor invasion and increased lymph node metastasis, with overexpression of ClC-3 being regulated by x-ray repair cross complementing 5 (68). ClC-3 is also significantly overexpressed in HCC, and its upregulation is associated with tumor size and overall prognosis, suggesting that ClC-3 expression may serve as a predictor of tumor size and overall prognosis in HCC (69). Furthermore, ClC-3 has been reported to be expressed in intestinal tissues and to be involved in the regulation of intestinal inflammation (70). ClC-3 deficiency may exacerbate inflammatory bowel disease (IBD) by promoting the apoptosis of intestinal epithelial cells and causing Paneth cell deficiency (Fig. 3), indicating that modulating ClC-3 expression could be a novel therapeutic strategy for treating IBD (71).
ClC-3 to ClC-7 are located primarily in intracellular organelles, particularly along the endosome-lysosome pathway, and are distributed differentially across compartments (43,72-74). ClC-4 localizes to various endosomal clusters, whereas ClC-5 is predominantly expressed in the kidney, where it localizes to early endosomes and participates in endocytic uptake in the proximal tubule. Both ClC-3 and ClC-4 are widely expressed and found in various endosomal populations. ClC-6 is expressed primarily in neurons and localizes to late endosomes, whereas ClC-7, along with its β-subunit osteopetrosis-associated transmembrane protein 1, localizes to lysosomes in all cell types and is also found at the ruffled borders of bone-resorbing osteoclasts. Dysfunction of ClC-3, ClC-4, ClC-6 and ClC-7 has been linked to intellectual disability, epilepsy, lysosomal storage disorders and neurodegeneration, respectively (75-77). However, to the best of our knowledge, their roles in digestive disorders have not yet been reported.
CFTR is a member of the ABC transporter protein superfamily (78) and functions as an anion channel. The anion-selective pore of CFTR is formed by two transmembrane domains (TMDs) regulated by cytoplasmic domains, which include two nucleotide-binding domains (NBDs) and a regulatory (R) domain. Channel activation requires cAMP-dependent protein kinase phosphorylation of the R domain, and the opening and closing (gating) of phosphorylated channels is driven by ATP binding and hydrolysis at NBDs (79).
Mutations in the CFTR gene lead to CF due to CFTR dysfunction. This gene encodes an anion channel that mediates the transport of chloride (Cl−) and bicarbonate (HCO3−) across epithelial surfaces, which is crucial for osmotic homeostasis and the maintenance of normal electrolyte transport in the lungs, upper respiratory tract, pancreas, liver, gallbladder, intestine and other exocrine glands on the apical surface of epithelial cells (80). While the CFTR protein is located primarily in the plasma membrane, increasing evidence suggests its presence in intracellular organelles such as endosomes, lysosomes, phagosomes and mitochondria. Dysfunction of CFTR not only impairs ion transport across epithelial tissues but also disrupts the normal functioning of these organelles (81).
Following CFTR dysfunction, bicarbonate ion transport is inhibited, disrupting the pH balance of the esophageal epithelium. This imbalance can result in cellular stress, DNA damage and increased susceptibility to carcinogenesis. In esophageal squamous cell carcinoma (ESCC), overexpression of CFTR was found to activate the p38 signaling pathway, which inhibits cell proliferation, migration and invasion; induces apoptosis; and is associated with a favorable patient prognosis (82). In esophageal cancer, gamma-aminobutyric acid receptor subunit pi(GABRP) promoted CFTR expression, and CFTR knockdown significantly counteracted the inhibitory effects of GABRP overexpression on EC cell proliferation, migration and invasion. These findings suggest that GABRP overexpression inhibits EC progression by increasing CFTR expression (83). Therefore, CFTR may act as a mediator and/or biomarker for ESCC (82). In addition, CFTR has been shown to inhibit the growth and migration of ESCC by downregulating NF-κB protein expression (23). CFTR modulators improve gastroesophageal reflux disease symptoms in patients with advanced CF (84). One of the specific reasons for this is that CFTR modulators improve gastroduodenal pH (85), in addition, CFTR is expressed in esophageal tissues and CFTR modulators may have a direct effect on esophageal motility and gastric emptying (82).
Pancreatitis is a common cause of hospitalization for digestive diseases, with high morbidity and mortality rates (86). It is a complex inflammatory disease of the acinar and ductal epithelium that is often caused by the premature activation of digestive enzymes (87). There is substantial evidence that CFTR plays a critical role as an anion transporter in maintaining normal pancreatic exocrine function. Dysfunction of CFTR triggers pancreatic disorders, including impaired insulin regulation, pancreatic ductal obstruction, acute and chronic inflammation and carcinogenesis (88,89). However, the specific pathophysiological mechanisms remain to be fully understood.
In the pancreas, digestive enzymes play a well-known role; however, electrolytes are equally important as carriers, helping to transport enzymes to the small intestine, neutralize gastric acid and increase the duodenal pH to the optimal level for enzyme activity (90). In particular, bicarbonate (HCO3−) secretion is essential, as it regulates the pH of the ductal epithelium, increases mucin secretion (which acts as an antimicrobial) and prevents the premature activation of pancreatic enzymes (91). CFTR is a key player in HCO3− secretion in pancreatic fluid. This secretion involves the coordinated action of ion-transporting proteins located on the basolateral and luminal membranes of pancreatic ductal cells (PDCs). HCO3− can accumulate via electrogenic sodium-bicarbonate cotransporter 1 (NBCe1), the solute carrier family 4 member 4 (SLC4A4) cotransporter protein, which is produced intracellularly through the carbonic anhydrase-catalyzed reaction of CO2 and water. It is then secreted into the ductal lumen via the coordinated action of CFTR and SLC26A6 (91,92). CFTR not only functions in conjunction with SLC26A6 but its R domain also binds to the spread through air spaces domain of SLC26A6, further regulating HCO3− secretion (93,94). CFTR channels directly mediate epithelial HCO3− transport and selectively secrete HCO3− when intracellular Cl− levels are low and HCO3− levels are relatively high (95). The dynamic regulation of CFTR anion conductance is thought to be mediated by With No Lysine Kinase (WNK) (18). WNK1, for example, has been shown to activate oxidative stress-responsive kinase 1 and STE20/SPS1-related proline/alanine-rich kinase (SPAK) at low extracellular Cl− concentrations, significantly increasing HCO3− secretion in CFTR-transfected 293 cells and guinea pig PDCs (96). However, WNK1 and WNK4 can also inhibit CFTR by reducing its level at the cell surface, thus inhibiting CFTR-mediated anion efflux (97). This inhibition is counteracted by the binding of the inositol 1,4,5-trisphosphate (IP3) receptor, with released IP3 antagonizing the WNK/SPAK pathway (98). IRBIT, through its PDZ domain, binds to CFTR and NBCe1 at the apical and basolateral membranes, recruiting protein phosphatase 1 to counteract SPAK-mediated phosphorylation of CFTR and NBCe1 (99).
IRBIT recruitment may involve signaling pathways involving cAMP and Ca2+ (100). Furthermore, CFTR is closely linked to the organization of the actin cytoskeleton and the formation of cellular junctions among epithelial cells, which are essential for establishing apical-basolateral polarity and ensuring proper epithelial barrier function. In CF cells, this barrier function is disrupted, suggesting that CF may not only be an ion channel disease but also a disorder of epithelial differentiation (101).
Studies have reported that Helicobacter pylori infection damages duodenal mucosal CFTR, contributing to the development of duodenal ulcers (102). The gastrointestinal tract relies on mucin production, maturation and transport to defend against pathogens and lubricate the epithelium. Research by Reyes et al (103) revealed that TNF facilitates ion transport and intestinal luminal flow via CFTR and that TNF deficiency leads to increased mucus accumulation in the gastrointestinal tract and reduced intestinal luminal fluid pumping, which slows intestinal transit. These findings suggest the existence of a TNF/CFTR signaling axis in the adult intestine, with epithelial cell-derived TNF identified as an upstream regulator of mucin homeostasis (103).
In addition, patients with CF are at increased risk of developing various types of cancer, particularly gastrointestinal cancers (104). A study involving 1,468 heterozygous germline carriers of the CFTR F508del mutation revealed an increased risk of GC in this population. Another report showed that serum CFTR levels in patients with GC correlated with the expression of the tumor biomarker CA199 (105). A study by Yamada et al (106) further highlighted that patients with CF have a significantly increased risk of gastrointestinal cancers, including small bowel cancer, and recommended the development of individualized screening protocols for site-specific gastrointestinal cancers in this population.
CFTR is expressed throughout the intestine, with a decreasing gradient of expression from the proximal (duodenum) to the distal (ileum) small intestine. In both the small and large intestines, CFTR is most strongly expressed at the base of the crypts and can influence intestinal stem cell function. Studies have demonstrated that CFTR is expressed by intestinal stem cells (107). The transmembrane domains of CFTR form water channels that allow Cl− and HCO3− ions to be secreted from epithelial cells into the intestinal lumen, particularly within the crypts, while simultaneously increasing water secretion in the same direction. This regulates the water homeostasis and pH of the intestine (108). CFTR also influences the composition of the intestinal flora (109,110), the maintenance of the epithelial barrier (111,112), and the balance of innate and adaptive immune responses (113,114). Clinical manifestations of gastrointestinal issues in patients with CF, such as inflammation, chronic abdominal pain and complications such as Crohn's disease and gastroesophageal reflux disease (115,116), are associated with dysregulation of these processes, increasing the risk of cancer. A 20-year epidemiologic study reported that patients with CF have an elevated risk of gastrointestinal cancers with age, particularly a sixfold increase in the risk of Colorectal Cancer (CRC), the most common gastrointestinal malignancy in patients with CF (117).
CFTR deficiency has also been linked to CRC in the general population, with disease-free survival being lower in patients with CRC with reduced CFTR expression in the tumor (118). CFTR-depleted CRC cell lines exhibit enhanced oncogenic features, including increased colony formation, migration and invasion (119). Increased β-catenin activity, associated with increased proliferation, has been observed in CFTR knockout crypt cells and organoids. Although studies on CRC without CF are limited, Palma et al (120) reported that CFTR may play a nontumor suppressive role in CRC initiation and progression by enhancing a set of genes associated with cancer stemness in patients without CFTR mutations. Mechanistic studies have shown that CFTR knockdown or transient inhibition by CFTR (inh)-172 leads to an increase in the intracellular pH of leucine-rich repeat-containing g-protein coupled receptor 5+ stem cells, promoting the binding of dishevelled segment polarity protein 2 (a member of the Wnt/β-catenin signaling pathway) to plasma membrane phospholipids and thereby enhancing Wnt/β-catenin signaling (121).
CFTR expression is downregulated in CRC, potentially due to promoter methylation. Overexpression of CFTR has been shown to inhibit CRC tumor growth by suppressing cell proliferation, migration and invasion. Furthermore, CFTR promoter methylation is significantly correlated with lymph node metastasis, suggesting that CFTR may serve as a potential marker for lymph node metastasis in patients with CRC (122).
CaCCs are chloride channels that can be synergistically activated by intracellular calcium and voltage. ANO1/TMEM16A contains 8 transmembrane structural domains (TM1-TM8), of which TM3-TM7 form the ion-conducting pore. The calcium binding site is located on the cytoplasmic side and contains multiple acidic residues responsible for calcium ion binding and channel activation. ANO1 exists as a dimer, with each monomer having a separate ion-conducting pore (123,124). CaCCs are expressed in both invertebrates and mammals, suggesting that they play crucial physiological roles, including regulating epithelial Cl− secretion, neuronal and cardiomyocyte excitability, smooth muscle contraction and injury perception (125). The main CaCCs identified so far are ANO1, also known as TMEM16A, and ANO2, also known as TMEM16B (126). ANO1 is widely expressed in various cancers, where it controls cancer cell proliferation, survival and migration (127). It also regulates the secretory function of epithelial cells (128) and the electrical pacing activity of interstitial cells of Cajal in gastrointestinal smooth muscle (129). By contrast, ANO2 (TMEM16B), although functioning as a CaCC, is expressed primarily in olfactory sensory neurons (130), hippocampal pyramidal neurons (131) and thalamocortical neurons in the brain (132).
Vanoni et al (133) investigated the relationship between eosinophilic esophagitis (EoE) and ANO1 and reported increased expression of ANO1 in the basal region of the esophagus in both esophageal biopsy samples from patients with EoE and from mice with EoE. This increase was positively correlated with disease severity. ANO1 serves as the primary apical Cl− transporter induced by IL-13 in the esophageal epithelium and regulates basal cell proliferation, a process linked to the regulation of TP63 expression and phosphorylated cell cycle protein-dependent kinase 2 (p-CDK2) levels (133). Additionally, a genome-wide copy number variation analysis revealed that ANO1 promotes ESCC cell proliferation, migration and invasion by activating the TGF-β pathway (Fig. 3). Knockdown of ANO1 significantly inhibited tumor progression both in vitro and in vivo, suggesting that ANO1 is a novel oncogene in ESCC and may serve as a prognostic biomarker for this disease (134).
ANO1 (TMEM16A) is the most well-known CaCC with typical Ca2+ and voltage-dependent activation, and it is regulated by phosphatidylinositol 4,5-bisphosphate (135). ANO1 has been associated with various gastrointestinal disorders. The interstitial cells of Cajal (Fig. 3), which act as pacemakers controlling smooth muscle contraction, express TMEM16A. The activation of TMEM16A is thought to promote gastrointestinal muscle contraction. Keratin is a TMEM16A activator that promotes contraction of smooth muscle cells (129), providing a theoretical basis for treating gastrointestinal dysfunction. ANO1 also regulates gastrointestinal motility (136), and mutations in this gene are linked to conditions such as diverticulitis, diverticulosis, congenital megacolon and constipation, likely due to its role in gastrointestinal pacemaker activity (137,138). ANO1 is highly expressed in gastrointestinal cancers and is correlated with tumor-lymph node-metastasis staging in patients with GC. Studies have shown that the knockdown of ANO1 significantly inhibits GC cell migration, invasion and metastasis in vitro. This involves a mechanism in which specificity protein 1 increases ANO1 transcription, recruits mixed lineage leukemia protein 1 to the ANO1 promoter region and promotes H3K4 trimethylation, leading to increased ANO1 expression (139). Additionally, opa interacting protein 5 antisense RNA 1 has been identified as a critical regulator of GC pathogenesis, acting as an oncogenic competitive endogenous RNA by binding and sequestering microRNA-422a to increase ANO1 expression (140). STAT6/ANO1 activation has also been shown to reduce the proliferation, migration and invasion of GC cells (141). In CRC, ANO1 expression is upregulated and is associated with the TNM stage, histologic type, pathological differentiation and poor prognosis. Knockdown of endogenous ANO1 inactivates the Wnt/β-catenin signaling pathway by downregulating key components (e.g., Frizzled protein 1 and β-catenin) and upregulating glycogen synthase kinase 3β, suggesting that targeting ANO1 may have therapeutic potential in colon cancer (142).
In research on acute pancreatitis (AP), IL-6 was found to promote TMEM16A (ANO1) expression via IL-6R/STAT3 signaling activation, and overexpression of TMEM16A further increased IL-6 secretion through the inositol 1,4,5-triphosphate receptor/Ca2+/NF-κB signaling pathway (143). Therefore, inhibiting TMEM16A could represent a novel therapeutic strategy for treating AP. In addition, studies have identified ANO1 as a prognostic factor after radical resection of pancreatic carcinoma, where ANO1 may induce an immunosuppressive tumor microenvironment in a paracrine manner (144), suggesting that ANO1 could be a potential therapeutic target for pancreatic cancer.
Ion channels have been extensively studied in liver diseases in recent years, but chloride channels, including TMEM16A (ANO1), have been less frequently explored. Studies have shown that TMEM16A expression is elevated in the liver tissues of both mice and patients with NAFLD. Hepatocyte-specific knockdown of TMEM16A in mice improved hepatic glucose metabolism disorders and steatosis. TMEM16A in hepatocytes interacts with vesicle-associated membrane protein 3 (VAMP3), inducing its degradation and inhibiting the formation of VAMP3/synaptobrevin complexes, such as VAMP3/synaptosome-associated protein 23 and VAMP3/syntaxin 4. This interaction disrupts the translocation of hepatic glucose transporter protein 2 (GLUT2), leading to impaired glucose uptake. Notably, VAMP3 overexpression counteracts the inhibitory effect of TMEM16A on GLUT2 translocation and helps reduce lipid deposition, insulin resistance and inflammation (145). These findings suggest that inhibiting hepatic TMEM16A or disrupting the TMEM16A/VAMP3 interaction could offer new therapeutic strategies for NAFLD. Guo et al (146) reported that the TMEM16A-glutathione peroxidase 4 (GPX4) interaction and GPX4 ubiquitination are essential for TMEM16A-regulated hepatic ischemia/reperfusion injury, suggesting that blockade of the TMEM16A-GPX4 interaction or inhibition of TMEM16A in hepatocytes may represent promising therapeutic strategies for acute liver injury. Kondo et al (147) reported that downregulation of TMEM16A expression in cirrhotic portal hypertension-associated portal vein smooth muscle cells reduces Ca2+-activated Cl− channel activity; it is mediated by the increase in angiotensin II in cirrhosis. In addition, while ANO1 plays a regulatory role in various cancers, its role in HCC has been less studied. TMEM16A is overexpressed in HCC, and the inhibition of TMEM16A suppresses the MAPK signaling pathway (Fig. 3), reducing HCC growth. These findings indicate that TMEM16A may serve as a promising therapeutic target for HCC (148).
VRACs, also known as volume-sensitive outwardly rectifying channels, regulate vertebrate cell volume by mediating the extrusion of chloride ions and organic osmolytes. VRACs are composed of a heterodimer of leucine-rich repeat-containing 8 (LRRC8) proteins, where LRRC8A (also known as SWELL1) serves as an essential subunit. LRRC8A binds to any of its homologs, such as LRRC8B-E, to form the hexameric VRACs complex, which functions as an anion-selective channel. VRACs are activated either by cellular swelling or by reactive oxygen species, a process dependent on intracellular ATP (149-151). VRACs play a crucial role in various physiological processes, including cell volume regulation, proliferation, migration and apoptosis (152).
A study by Konishi et al (153) revealed that high expression of LRRC8A in ESCC tissues from patients was associated with poor prognosis. LRRC8A mediated the proliferation, survival and migration of ESCC cell lines. It regulates the expression of genes involved in the G1/S cell cycle checkpoint, including p21, p27 and E2F7, as well as genes related to cell migration, such as matrix metalloproteinase 1 and integrin αV. Depletion of LRRC8A reduced cell proliferation and migration and promoted apoptosis, and microarray analysis demonstrated that LRRC8A depletion inhibited the PI3K/AKT signaling pathway in cells (153).
LRRC8A has been shown to influence the growth of GC cells, and high expression of LRRC8A was identified as an independent prognostic factor for 5-year survival in patients with GC on the basis of multivariate analysis of clinical samples. Further research revealed that knockdown of LRRC8A inhibited cell proliferation and migration, enhanced apoptosis and affected the expression of genes associated with the p53 signaling pathway, including JNK, p53, p21, Bcl-2 and FAS (154).
Pancreatic adenocarcinoma (PAAD) is a highly malignant tumor of the digestive system with increasing morbidity and mortality rates. LRRC8A has been shown to influence the prognosis of PAAD and to be associated with cell proliferation, migration, drug resistance and immune infiltration. Notably, LRRC8A plays a role in the progression and prognosis of patients with PAAD by regulating the immune infiltration of CD8+ T cells (155).
SWELL1, an integral component of the VRACs, is highly expressed in HCC tissues and is associated with poor prognosis. An in vitro study showed that the overexpression of SWELL1 significantly promoted cell proliferation and migration while inhibiting apoptosis. Mechanistically, SWELL1 interacts with protein kinase Cα, activates the cyclin D1/cyclin-dependent kinase 2 pathway to induce cell growth, and regulates cell migration through the JNK pathway (156).
LRRC8A was found to be elevated in 60% of CRC patient tissues, and patients with high LRRC8A expression had shorter survival times, suggesting that LRRC8A may serve as a novel prognostic biomarker for CRC. Elevated expression of LRRC8A has been linked to enhanced cancer cell growth and metastasis (157), whereas downregulation of VRACs has been shown to inhibit colon cancer cell proliferation (158). Furthermore, one study reported that LRRC8A contributed to oxaliplatin resistance in colon cancer cells (159). Of note, exosomes, which are crucial mediators of intercellular communication in the tumor microenvironment and accelerate colon cancer progression, contain LRRC8A. This protein, which is one of the components of exosomes released from colon cancer HCT116 cells, is responsible for exosome production and is involved in volume regulation (160).
PACs, also known as acid-sensitive outwardly rectifying anion channels (161), are assembled from symmetric trimers of TMEM206. Each subunit consists of a large extracellular domain (ECD), which resides in the lumen of endosomes, and two TMDs with helices (TM1 and TM2) connected by a near-membrane interface that links the ECD to the TMD. Acidic clusters in the ECD serve as primary proton sensors under acidic conditions (162).
Research has reported elevated TMEM206 expression in CRC tissues compared with adjacent noncancer tissues. TMEM206 promotes CRC cell proliferation, invasion and migration by increasing the levels of phosphorylated AKT and downstream signaling components, as well as the level of phosphorylated extracellular signal-regulated kinase (ERK), facilitating interactions between the AKT and ERK pathways (163). Bioinformatics analysis revealed that TMEM206 expression is significantly elevated in CRC, breast cancer, HCC and lymphoma tissues compared with corresponding normal tissues, identifying TMEM206 as a potential prognostic biomarker for HCC (164).
Ligand-gated chloride channels, such as GABA-A and glycine receptor channels, consist of five subunits that form the central ionotropic pore. Each subunit contains a characteristic Cys-loop structural domain involved in ligand binding and channel gating (165). They primarily play roles in the development and function of the nervous system (166). Channelopathies involving these channels have been linked to neurological disorders such as Parkinson's disease, Huntington's disease and Alzheimer's disease (167-169). However, to the best of our knowledge, no studies have explored the role of these channels in digestive disorders.
Ion channels and transport proteins play crucial roles in regulating pathophysiological processes across various organs of the digestive system. In recent years, sodium, calcium and potassium ion channels have been extensively studied, whereas chloride channels have received comparatively less attention. Chloride, the most abundant anion in the body, is regulated primarily by chloride channels or transporters. Existing studies on chloride channels have demonstrated their significant role in the development of numerous digestive system diseases (Table I), including esophagitis, esophageal cancer, gastrointestinal dyskinesia, gastritis, GC, colonic diverticulitis, diverticulosis, congenital megacolon, colon cancer, pancreatitis, pancreatic cancer, hepatic lipid metabolism disorders and HCC. Although the involvement of chloride channels in the digestive system has been explored, the underlying physiological and pathological mechanisms within various organs have not been thoroughly investigated. Most research to date has focused on verifying the high or low expression of chloride channels in digestive diseases or their involvement in relevant signaling pathways, whereas fewer studies have delved into deeper mechanisms such as transcriptional regulation or posttranslational modifications. Therefore, further investigations into chloride channels constitute an important and promising research area that remains to be fully explored.
Not applicable.
YH wrote the original manuscript and performed the literature review. BT was responsible for the conceptualization and review of the study and provided supervision. Both authors have read and approved the final version of the manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
The authors thank Professor Hui Dong (Department of Medicine, School of Medicine, University of California, San Diego University, San Diego, USA) for providing guidance in the writing of the paper.
The authors gratefully acknowledge financial support from the National Natural Science Foundation of China (grant no. 81960507), the Basic Research Projects of Science and Technology Department of Guizhou Province (grant code, Qian Ke He-zk[2022]-646) and the 2023 Graduate Research Fund Program B (grant no. ZYK248).
|
Nguyen HX and Bursac N: Ion channel engineering for modulation and de novo generation of electrical excitability. Curr Opin Biotechnol. 58:100–107. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Capatina AL, Lagos D and Brackenbury WJ: Targeting ion channels for cancer treatment: Current progress and future challenges. Rev Physiol Biochem Pharmacol. 183:1–43. 2022. | |
|
Bulk E, Todesca LM and Schwab A: Ion channels in lung cancer. Rev Physiol Biochem Pharmacol. 181:57–79. 2021. View Article : Google Scholar | |
|
Xiao F, Yu Q, Li J, Johansson ME, Singh AK, Xia W, Riederer B, Engelhardt R, Montrose M, Soleimani M, et al: Slc26a3 deficiency is associated with loss of colonic HCO3 (-) secretion, absence of a firm mucus layer and barrier impairment in mice. Acta Physiol (Oxf). 211:161–175. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Zahn A, Moehle C, Langmann T, Ehehalt R, Autschbach F, Stremmel W and Schmitz G: Aquaporin-8 expression is reduced in ileum and induced in colon of patients with ulcerative colitis. World J Gastroenterol. 13:1687–1695. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Xu H, Zhang B, Li J, Wang C, Chen H and Ghishan FK: Impaired mucin synthesis and bicarbonate secretion in the colon of NHE8 knockout mice. Am J Physiol Gastrointest Liver Physiol. 303:G335–G343. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Singh AK, Sjöblom M, Zheng W, Krabbenhöft A, Riederer B, Rausch B, Manns MP, Soleimani M and Seidler U: CFTR and its key role in in vivo resting and luminal acid-induced duodenal HCO3-secretion. Acta Physiol (Oxf). 193:357–365. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Schultheis PJ, Clarke LL, Meneton P, Harline M, Boivin GP, Stemmermann G, Duffy JJ, Doetschman T, Miller ML and Shull GE: Targeted disruption of the murine Na+/H+ exchanger isoform 2 gene causes reduced viability of gastric parietal cells and loss of net acid secretion. J Clin Invest. 101:1243–1253. 1998. View Article : Google Scholar : PubMed/NCBI | |
|
Wang A, Li J, Zhao Y, Johansson ME, Xu H and Ghishan FK: Loss of NHE8 expression impairs intestinal mucosal integrity. Am J Physiol Gastrointest Liver Physiol. 309:G855–G864. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Boivin GP, Schultheis PJ, Shull GE and Stemmermann GN: Variant form of diffuse corporal gastritis in NHE2 knockout mice. Comp Med. 50:511–515. 2000.PubMed/NCBI | |
|
Ding X, Li D, Li M, Wang H, He Q, Wang Y, Yu H, Tian D and Yu Q: SLC26A3 (DRA) prevents TNF-alpha-induced barrier dysfunction and dextran sulfate sodium-induced acute colitis. Lab Invest. 98:462–476. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang W, Xu Y, Chen Z, Xu Z and Xu H: Knockdown of aquaporin 3 is involved in intestinal barrier integrity impairment. FEBS Lett. 585:3113–3119. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Moeser AJ, Nighot PK, Ryan KA, Simpson JE, Clarke LL and Blikslager AT: Mice lacking the Na+/H+ exchanger 2 have impaired recovery of intestinal barrier function. Am J Physiol Gastrointest Liver Physiol. 295:G791–G797. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Kumar A, Priyamvada S, Ge Y, Jayawardena D, Singhal M, Anbazhagan AN, Chatterjee I, Dayal A, Patel M, Zadeh K, et al: A novel role of SLC26A3 in the maintenance of intestinal epithelial barrier integrity. Gastroenterology. 160:1240–1255.e3. 2021. View Article : Google Scholar | |
|
Doi K, Nagao T, Kawakubo K, Ibayashi S, Aoyagi K, Yano Y, Yamamoto C, Kanamoto K, Iida M, Sadoshima S and Fujishima M: Calcitonin gene-related peptide affords gastric mucosal protection by activating potassium channel in Wistar rat. Gastroenterology. 114:71–76. 1998. View Article : Google Scholar : PubMed/NCBI | |
|
Deng Z, Zhao Y, Ma Z, Zhang M, Wang H, Yi Z, Tuo B, Li T and Liu X: Pathophysiological role of ion channels and transporters in gastrointestinal mucosal diseases. Cell Mol Life Sci. 78:8109–8125. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Pallagi P, Hegyi P and Rakonczay Z Jr: The physiology and pathophysiology of pancreatic ductal secretion: The background for clinicians. Pancreas. 44:1211–1233. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Ishiguro H, Yamamoto A, Nakakuki M, Yi L, Ishiguro M, Yamaguchi M, Kondo S and Mochimaru Y: Physiology and pathophysiology of bicarbonate secretion by pancreatic duct epithelium. Nagoya J Med Sci. 74:1–18. 2012.PubMed/NCBI | |
|
Novak I, Haanes KA and Wang J: Acid-base transport in pancreas-new challenges. Front Physiol. 4:3802013. View Article : Google Scholar | |
|
Schnipper J, Dhennin-Duthille I, Ahidouch A and Ouadid-Ahidouch H: Ion channel signature in healthy pancreas and pancreatic ductal adenocarcinoma. Front Pharmacol. 11:5689932020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen X, Zhang L, Zheng L and Tuo B: Role of Ca(2+) channels in non-alcoholic fatty liver disease and their implications for therapeutic strategies (Review). Int J Mol Med. 50:1132022. View Article : Google Scholar : PubMed/NCBI | |
|
Stock C: How dysregulated ion channels and transporters take a hand in esophageal, liver, and colorectal cancer. Rev Physiol Biochem Pharmacol. 181:129–222. 2021. View Article : Google Scholar | |
|
Li W, Wang C, Peng X, Zhang H, Huang H and Liu H: CFTR inhibits the invasion and growth of esophageal cancer cells by inhibiting the expression of NF-κB. Cell Biol Int. 42:1680–1687. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Goldman A, Chen H, Khan MR, Roesly H, Hill KA, Shahidullah M, Mandal A, Delamere NA and Dvorak K: The Na+/H+ exchanger controls deoxycholic acid-induced apoptosis by a H+-activated, Na+-dependent ionic shift in esophageal cells. PLoS One. 6:e238352011. View Article : Google Scholar | |
|
Boult J, Roberts K, Brookes MJ, Hughes S, Bury JP, Cross SS, Anderson GJ, Spychal R, Iqbal T and Tselepis C: Overexpression of cellular iron import proteins is associated with malignant progression of esophageal adenocarcinoma. Clin Cancer Res. 14:379–387. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Gao N, Yang F, Chen S, Wan H, Zhao X and Dong H: The role of TRPV1 ion channels in the suppression of gastric cancer development. Exp Clin Cancer Res. 39:2062020. View Article : Google Scholar | |
|
Chang Y, Roy S and Pan Z: Store-operated calcium channels as drug target in gastroesophageal cancers. Front Pharmacol. 12:6687302021. View Article : Google Scholar : PubMed/NCBI | |
|
Csekő K, Pécsi D, Kajtár B, Hegedűs I, Bollenbach A, Tsikas D, Szabó IL, Szabó S and Helyes Z: Upregulation of the TRPA1 Ion channel in the gastric mucosa after iodoacetamide-induced gastritis in rats: A potential new therapeutic target. Int J Mol Sci. 21:55912020. View Article : Google Scholar | |
|
Capurro MI, Greenfield LK, Prashar A, Xia S, Abdullah M, Wong H, Zhong XZ, Bertaux-Skeirik N, Chakrabarti J, Siddiqui I, et al: VacA generates a protective intracellular reservoir for Helicobacter pylori that is eliminated by activation of the lysosomal calcium channel TRPML1. Nat Microbiol. 4:1411–1423. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang L, Zhang Y, Jiang Y, Dou X, Li S, Chai H, Qian Q and Wang M: Upregulated SOCC and IP3R calcium channels and subsequent elevated cytoplasmic calcium signaling promote nonalcoholic fatty liver disease by inhibiting autophagy. Mol Cell Biochem. 476:3163–3175. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Swain SM, Romac JM, Shahid RA, Pandol SJ, Liedtke W, Vigna SR and Liddle RA: TRPV4 channel opening mediates pressure-induced pancreatitis initiated by Piezo1 activation. J Clin Invest. 130:2527–2541. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lin R, Bao X, Wang H, Zhu S, Liu Z, Chen Q, Ai K and Shi B: TRPM2 promotes pancreatic cancer by PKC/MAPK pathway. Cell Death Dis. 12:5852021. View Article : Google Scholar : PubMed/NCBI | |
|
Li S, Han J, Guo G, Sun Y, Zhang T, Zhao M, Xu Y, Cui Y, Liu Y and Zhang J: Voltage-gated sodium channels β3 subunit promotes tumorigenesis in hepatocellular carcinoma by facilitating p53 degradation. FEBS Lett. 594:497–508. 2020. View Article : Google Scholar | |
|
Nattramilarasu PK, Bücker R, Lobo de Sá FD, Fromm A, Nagel O, Lee IM, Butkevych E, Mousavi S, Genger C, Kløve S, et al: Campylobacter concisus impairs sodium absorption in colonic epithelium via ENaC dysfunction and claudin-8 disruption. Int J Mol Sci. 21:3732020. View Article : Google Scholar : PubMed/NCBI | |
|
Gao F, Wang D, Liu X, Wu YH, Wang HT and Sun SL: Sodium channel 1 subunit alpha SCNN1A exerts oncogenic function in pancreatic cancer via accelerating cellular growth and metastasis. Arch Biochem Biophys. 727:1093232022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Li C, Peng M, Wang L, Zhao D, Wu T, Yi D, Hou Y and Wu G: N-Acetylcysteine improves intestinal function and attenuates intestinal autophagy in piglets challenged with β-conglycinin. Sci Rep. 11:12612021. View Article : Google Scholar | |
|
Goswami S: Interplay of potassium channel, gastric parietal cell and proton pump in gastrointestinal physiology, pathology and pharmacology. Minerva Gastroenterol (Torino). 68:289–305. 2022. | |
|
Patel SH, Edwards MJ and Ahmad SA: Intracellular ion channels in pancreas cancer. Cell Physiol Biochem. 53:44–51. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Schroeder BC, Waldegger S, Fehr S, Bleich M, Warth R, Greger R and Jentsch TJ: A constitutively open potassium channel formed by KCNQ1 and KCNE3. Nature. 403:196–199. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Ponnalagu D and Singh H: Anion channels of mitochondria. Handb Exp Pharmacol. 240:71–101. 2017. View Article : Google Scholar | |
|
Gururaja Rao S, Ponnalagu D, Patel NJ and Singh H: Three decades of chloride intracellular channel proteins: From organelle to organ physiology. Curr Protoc Pharmacol. 80:11.21.1–11.21.17. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Kouyoumdzian NM, Kim G, Rudi MJ, Rukavina Mikusic NL, Fernández BE and Choi MR: Clues and new evidences in arterial hypertension: Unmasking the role of the chloride anion. Pflugers Arch. 474:155–176. 2022. View Article : Google Scholar | |
|
Jentsch TJ and Pusch M: CLC chloride channels and transporters: Structure, function, physiology, and disease. Physiol Rev. 98:1493–1590. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Shi J, Shi S, Yuan G, Jia Q, Shi S, Zhu X, Zhou Y, Chen T and Hu Y: Bibliometric analysis of chloride channel research (2004-2019). Channels (Austin). 14:393–402. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Jentsch TJ, Stein V, Weinreich F and Zdebik AA: Molecular structure and physiological function of chloride channels. Physiol Rev. 82:503–568. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Peng F, Wu Y, Dong X and Huang P: Proton-activated chloride channel: Physiology and disease. Front Biosci (Landmark Ed). 28:112023. View Article : Google Scholar : PubMed/NCBI | |
|
Patil VM and Gupta SP: Studies on chloride channels and their modulators. Curr Top Med Chem. 16:1862–1876. 2016. View Article : Google Scholar | |
|
Accardi A and Miller C: Secondary active transport mediated by a prokaryotic homologue of ClC Cl- hannels. Nature. 427:803–807. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Jentsch TJ: CLC chloride channels and transporters: From genes to protein structure, pathology and physiology. Crit Rev Biochem Mol Biol. 43:3–36. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Yang YD, Cho H, Koo JY, Tak MH, Cho Y, Shim WS, Park SP, Lee J, Lee B, Kim BM, et al: TMEM16A confers receptor-activated calcium-dependent chloride conductance. Nature. 455:1210–1215. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Caputo A, Caci E, Ferrera L, Pedemonte N, Barsanti C, Sondo E, Pfeffer U, Ravazzolo R, Zegarra-Moran O and Galietta LJ: TMEM16A, a membrane protein associated with calcium-dependent chloride channel activity. Science. 322:590–594. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Gadsby DC, Vergani P and Csanády L: The ABC protein turned chloride channel whose failure causes cystic fibrosis. Nature. 440:477–483. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Riordan JR, Rommens JM, Kerem B, Alon N, Rozmahel R, Grzelczak Z, Zielenski J, Lok S, Plavsic N, Chou JL, et al: Identification of the cystic fibrosis gene: Cloning and characterization of complementary DNA. Science. 245:1066–1073. 1989. View Article : Google Scholar : PubMed/NCBI | |
|
Ullrich F, Blin S, Lazarow K, Daubitz T, von Kries JP and Jentsch TJ: Identification of TMEM206 proteins as pore of PAORAC/ASOR acid-sensitive chloride channels. Elife. 8:e491872019. View Article : Google Scholar : PubMed/NCBI | |
|
Olsen RW and Sieghart W: International Union of Pharmacology. LXX. Subtypes of gamma-aminobutyric acid(A) receptors: Classification on the basis of subunit composition, pharmacology, and function. Update. Pharmacol Rev. 60:243–260. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Lynch JW: Molecular structure and function of the glycine receptor chloride channel. Physiol Rev. 84:1051–1095. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Peng JM, Lin SH, Yu MC and Hsieh SY: CLIC1 recruits PIP5K1A/C to induce cell-matrix adhesions for tumor metastasis. J Clin Invest. 131:e1335252021. View Article : Google Scholar : | |
|
Bi MM, Hong S, Zhou HY, Wang HW, Wang LN and Zheng YJ: Chloride channelopathies of ClC-2. Int J Mol Sci. 15:218–249. 2013. View Article : Google Scholar | |
|
Okada Y, Okada T, Sato-Numata K, Islam MR, Ando-Akatsuka Y, Numata T, Kubo M, Shimizu T, Kurbannazarova RS, Marunaka Y and Sabirov RZ: Cell volume-activated and volume-correlated anion channels in mammalian cells: Their biophysical, molecular, and pharmacological properties. Pharmacol Rev. 71:49–88. 2019. View Article : Google Scholar | |
|
Fu D, Cui H and Zhang Y: Lack of ClC-2 alleviates high fat diet-induced insulin resistance and non-alcoholic fatty liver disease. Cell Physiol Biochem. 45:2187–2198. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Blikslager AT, Moeser AJ, Gookin JL, Jones SL and Odle J: Restoration of barrier function in injured intestinal mucosa. Physiol Rev. 87:545–564. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Gehren AS, Rocha MR, de Souza WF and Morgado-Díaz JA: Alterations of the apical junctional complex and actin cytoskeleton and their role in colorectal cancer progression. Tissue Barriers. 3:e10176882015. View Article : Google Scholar : PubMed/NCBI | |
|
Nighot PK, Moeser AJ, Ryan KA, Ghashghaei T and Blikslager AT: ClC-2 is required for rapid restoration of epithelial tight junctions in ischemic-injured murine jejunum. Exp Cell Res. 315:110–118. 2009. View Article : Google Scholar | |
|
Jin Y, Ibrahim D, Magness ST and Blikslager AT: Knockout of ClC-2 reveals critical functions of adherens junctions in colonic homeostasis and tumorigenicity. Am J Physiol Gastrointest Liver Physiol. 315:G966–G979. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Mrad FCC, Soares SBM, de Menezes Silva LAW, Dos Anjos Menezes PV and Simões-E-Silva AC: Bartter's syndrome: Clinical findings, genetic causes and therapeutic approach. World J Pediatr. 17:31–39. 2021. View Article : Google Scholar | |
|
Andrini O, Eladari D and Picard N: ClC-K kidney chloride channels: From structure to pathology. Handb Exp Pharmacol. 283:35–58. 2024. View Article : Google Scholar | |
|
Mu H, Mu L and Gao J: Suppression of CLC-3 reduces the proliferation, invasion and migration of colorectal cancer through Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun. 533:1240–1246. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Gu Z, Li Y, Yang X, Yu M, Chen Z, Zhao C, Chen L and Wang L: Overexpression of CLC-3 is regulated by XRCC5 and is a poor prognostic biomarker for gastric cancer. J Hematol Oncol. 11:1152018. View Article : Google Scholar : PubMed/NCBI | |
|
Cheng W, Zheng S, Li L, Zhou Q, Zhu H, Hu J and Luo H: Chloride channel 3 (CIC-3) predicts the tumor size in hepatocarcinoma. Acta Histochem. 121:284–288. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Huang LY, Li YJ, Li PP, Li HC and Ma P: Aggravated intestinal apoptosis by ClC-3 deletion is lethal to mice endotoxemia. Cell Biol Int. 42:1445–1453. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Huang LY, He Q, Liang SJ, Su YX, Xiong LX, Wu QQ, Wu QY, Tao J, Wang JP, Tang YB, et al: ClC-3 chloride channel/antiporter defect contributes to inflammatory bowel disease in humans and mice. Gut. 63:1587–1595. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Stauber T, Weinert S and Jentsch TJ: Cell biology and physiology of CLC chloride channels and transporters. Compr Physiol. 2:1701–1744. 2012. View Article : Google Scholar | |
|
Stauber T and Jentsch TJ: Chloride in vesicular trafficking and function. Annu Rev Physiol. 75:453–477. 2013. View Article : Google Scholar | |
|
Wartosch L, Fuhrmann JC, Schweizer M, Stauber T and Jentsch TJ: Lysosomal degradation of endocytosed proteins depends on the chloride transport protein ClC-7. FASEB J. 23:4056–4068. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Nicoli ER, Weston MR, Hackbarth M, Becerril A, Larson A, Zein WM, Baker PR II, Burke JD, Dorward H, Davids M, et al: Lysosomal storage and albinism due to effects of a de novo CLCN7 variant on lysosomal acidification. Am J Hum Genet. 104:1127–1138. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Polovitskaya MM, Barbini C, Martinelli D, Harms FL, Cole FS, Calligari P, Bocchinfuso G, Stella L, Ciolfi A, Niceta M, et al: A recurrent gain-of-function mutation in CLCN6, encoding the ClC-6 Cl(-)/H(+)-exchanger, causes early-onset neurodegeneration. Am J Hum Genet. 107:1062–1077. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Bose S, He H and Stauber T: Neurodegeneration upon dysfunction of endosomal/lysosomal CLC chloride transporters. Front Cell Dev Biol. 9:6392312021. View Article : Google Scholar : PubMed/NCBI | |
|
Locher KP: Mechanistic diversity in ATP-binding cassette (ABC) transporters. Nat Struct Mol Biol. 23:487–493. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Csanády L, Vergani P and Gadsby DC: Structure, gating, and regulation of the CFTR anion channel. Physiol Rev. 99:707–738. 2019. View Article : Google Scholar | |
|
Fonseca C, Bicker J, Alves G, Falcão A and Fortuna A: Cystic fibrosis: Physiopathology and the latest pharmacological treatments. Pharmacol Res. 162:1052672020. View Article : Google Scholar : PubMed/NCBI | |
|
Lukasiak A and Zajac M: The distribution and role of the CFTR protein in the intracellular compartments. Membranes (Basel). 11:8042021. View Article : Google Scholar : PubMed/NCBI | |
|
Matsumoto Y, Shiozaki A, Kosuga T, Kudou M, Shimizu H, Arita T, Konishi H, Komatsu S, Kubota T, Fujiwara H, et al: Expression and role of CFTR in human esophageal squamous cell carcinoma. Ann Surg Oncol. 28:6424–6436. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang J, Liu X, Zeng L and Hu Y: GABRP inhibits the progression of oesophageal cancer by regulating CFTR: Integrating bioinformatics analysis and experimental validation. Int J Exp Pathol. 105:118–132. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Shakir S, Echevarria C, Doe S, Brodlie M, Ward C and Bourke SJ: Elexacaftor-tezacaftor-ivacaftor improve gastro-oesophageal reflux and sinonasal symptoms in advanced cystic fibrosis. J Cyst Fibros. 21:807–810. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Rowe SM, Heltshe SL, Gonska T, Donaldson SH, Borowitz D, Gelfond D, Sagel SD, Khan U, Mayer-Hamblett N, Van Dalfsen JM, et al: Clinical mechanism of the cystic fibrosis transmembrane conductance regulator potentiator ivacaftor in G551D-mediated cystic fibrosis. Am J Respir Crit Care Med. 190:175–184. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Petrov MS and Yadav D: Global epidemiology and holistic prevention of pancreatitis. Nat Rev Gastroenterol Hepatol. 16:175–184. 2019. View Article : Google Scholar : | |
|
Mayerle J, Sendler M, Hegyi E, Beyer G, Lerch MM and Sahin-Tóth M: Genetics, cell biology, and pathophysiology of pancreatitis. Gastroenterology. 156:1951–1968.e1. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Hou Y, Guan X, Yang Z and Li C: Emerging role of cystic fibrosis transmembrane conductance regulator-an epithelial chloride channel in gastrointestinal cancers. World J Gastrointest Oncol. 8:282–288. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Fűr G, Bálint ER, Orján EM, Balla Z, Kormányos ES, Czira B, Szűcs A, Kovács DP, Pallagi P, Maléth J, et al: Mislocalization of CFTR expression in acute pancreatitis and the beneficial effect of VX-661 + VX-770 treatment on disease severity. J Physiol. 599:4955–4971. 2021. View Article : Google Scholar | |
|
Kim Y, Jun I, Shin DH, Yoon JG, Piao H, Jung J, Park HW, Cheng MH, Bahar I, Whitcomb DC and Lee MG: Regulation of CFTR bicarbonate channel activity by WNK1: Implications for pancreatitis and CFTR-Related disorders. Cell Mol Gastroenterol Hepatol. 9:79–103. 2020. View Article : Google Scholar | |
|
Lee MG, Ohana E, Park HW, Yang D and Muallem S: Molecular mechanism of pancreatic and salivary gland fluid and HCO3 secretion. Physiol Rev. 92:39–74. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Fong P: CFTR-SLC26 transporter interactions in epithelia. Biophys Rev. 4:107–116. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Ko SB, Zeng W, Dorwart MR, Luo X, Kim KH, Millen L, Goto H, Naruse S, Soyombo A, Thomas PJ and Muallem S: Gating of CFTR by the STAS domain of SLC26 transporters. Nat Cell Biol. 6:343–350. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Berg P, Svendsen SL, Sorensen MV, Larsen CK, Andersen JF, Jensen-Fangel S, Jeppesen M, Schreiber R, Cabrita I, Kunzelmann K and Leipziger J: Impaired renal HCO(3)(-) excretion in cystic fibrosis. J Am Soc Nephrol. 31:1711–1727. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Ishiguro H, Steward MC, Naruse S, Ko SB, Goto H, Case RM, Kondo T and Yamamoto A: CFTR functions as a bicarbonate channel in pancreatic duct cells. J Gen Physiol. 133:315–326. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Park HW, Nam JH, Kim JY, Namkung W, Yoon JS, Lee JS, Kim KS, Venglovecz V, Gray MA, Kim KH and Lee MG: Dynamic regulation of CFTR bicarbonate permeability by [Cl-]i and its role in pancreatic bicarbonate secretion. Gastroenterology. 139:620–631. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Yang CL, Liu X, Paliege A, Zhu X, Bachmann S, Dawson DC and Ellison DH: WNK1 and WNK4 modulate CFTR activity. Biochem Biophys Res Commun. 353:535–540. 2007. View Article : Google Scholar | |
|
Yang D, Li Q, So I, Huang CL, Ando H, Mizutani A, Seki G, Mikoshiba K, Thomas PJ and Muallem S: IRBIT governs epithelial secretion in mice by antagonizing the WNK/SPAK kinase pathway. J Clin Invest. 121:956–965. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Shirakabe K, Priori G, Yamada H, Ando H, Horita S, Fujita T, Fujimoto I, Mizutani A, Seki G and Mikoshiba K: IRBIT, an inositol 1,4,5-trisphosphate receptor-binding protein, specifically binds to and activates pancreas-type Na+/HCO3-cotransporter 1 (pNBC1). Proc Natl Acad Sci USA. 103:9542–9547. 2006. View Article : Google Scholar | |
|
Park S, Shcheynikov N, Hong JH, Zheng C, Suh SH, Kawaai K, Ando H, Mizutani A, Abe T, Kiyonari H, et al: Irbit mediates synergy between ca(2+) and cAMP signaling pathways during epithelial transport in mice. Gastroenterology. 145:232–241. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Pankonien I, Quaresma MC, Rodrigues CS and Amaral MD: CFTR, cell junctions and the cytoskeleton. Int J Mol Sci. 23:2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wen G, Jin H, Deng S, Xu J, Liu X, Xie R and Tuo B: Effects of helicobacter pylori infection on the expressions and functional activities of human duodenal mucosal bicarbonate transport proteins. Helicobacter. 21:536–547. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Reyes EA, Castillo-Azofeifa D, Rispal J, Wald T, Zwick RK, Palikuqi B, Mujukian A, Rabizadeh S, Gupta AR, Gardner JM, et al: Epithelial TNF controls cell differentiation and CFTR activity to maintain intestinal mucin homeostasis. J Clin Invest. 133:e1635912023. View Article : Google Scholar : PubMed/NCBI | |
|
Bhattacharya R, Blankenheim Z, Scott PM and Cormier RT: CFTR and gastrointestinal cancers: An update. J Pers Med. 12:8682022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu H, Wu W, Liu Y, Zhang C and Zhou Z: Predictive value of cystic fibrosis transmembrane conductance regulator (CFTR) in the diagnosis of gastric cancer. Clinical and investigative medicine. Clin Invest Med. 37:E226–E232. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Yamada A, Komaki Y, Komaki F, Micic D, Zullow S and Sakuraba A: Risk of gastrointestinal cancers in patients with cystic fibrosis: A systematic review and meta-analysis. Lancet Oncol. 19:758–767. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Jakab RL, Collaco AM and Ameen NA: Physiological relevance of cell-specific distribution patterns of CFTR, NKCC1, NBCe1, and NHE3 along the crypt-villus axis in the intestine. Am J Physiol Gastrointest Liver Physiol. 300:G82–G98. 2011. View Article : Google Scholar : | |
|
Tse CM, Yin J, Singh V, Sarker R, Lin R, Verkman AS, Turner JR and Donowitz M: cAMP stimulates SLC26A3 activity in human colon by a CFTR-dependent mechanism that does not require CFTR activity. Cell Mol Gastroenterol Hepatol. 7:641–653. 2019. View Article : Google Scholar : | |
|
Vernocchi P, Del Chierico F, Russo A, Majo F, Rossitto M, Valerio M, Casadei L, La Storia A, De Filippis F, Rizzo C, et al: Gut microbiota signatures in cystic fibrosis: Loss of host CFTR function drives the microbiota enterophenotype. PLoS One. 13:e02081712018. View Article : Google Scholar : PubMed/NCBI | |
|
Burke DG, Fouhy F, Harrison MJ, Rea MC, Cotter PD, O'Sullivan O, Stanton C, Hill C, Shanahan F, Plant BJ and Ross RP: The altered gut microbiota in adults with cystic fibrosis. BMC Microbiol. 17:582017. View Article : Google Scholar : PubMed/NCBI | |
|
De Lisle RC: Disrupted tight junctions in the small intestine of cystic fibrosis mice. Cell Tissue Res. 355:131–142. 2014. View Article : Google Scholar : | |
|
Broadbent D, Ahmadzai MM, Kammala AK, Yang C, Occhiuto C, Das R and Subramanian H: Roles of NHERF family of PDZ-binding proteins in regulating GPCR functions. Adv Immunol. 136:353–385. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Meeker SM, Mears KS, Sangwan N, Brittnacher MJ, Weiss EJ, Treuting PM, Tolley N, Pope CE, Hager KR, Vo AT, et al: CFTR dysregulation drives active selection of the gut microbiome. PLoS Pathog. 16:e10082512020. View Article : Google Scholar : PubMed/NCBI | |
|
Mulcahy EM, Cooley MA, McGuire H, Asad S, Fazekas de St Groth B, Beggs SA and Roddam LF: Widespread alterations in the peripheral blood innate immune cell profile in cystic fibrosis reflect lung pathology. Immunol Cell Biol. 97:416–426. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Trigo Salado C, Leo Carnerero E and de la Cruz Ramírez MD: Crohn's disease and cystic fibrosis: There is still a lot to learn. Rev Esp Enferm Dig. 110:835–836. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Gabel ME, Galante GJ and Freedman SD: Gastrointestinal and hepatobiliary disease in cystic fibrosis. Semin Respir Crit Care Med. 40:825–841. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Maisonneuve P, Marshall BC, Knapp EA and Lowenfels AB: Cancer risk in cystic fibrosis: A 20-year nationwide study from the United States. J Natl Cancer Inst. 105:122–129. 2013. View Article : Google Scholar | |
|
Than BL, Linnekamp JF, Starr TK, Largaespada DA, Rod A, Zhang Y, Bruner V, Abrahante J, Schumann A, Luczak T, et al: CFTR is a tumor suppressor gene in murine and human intestinal cancer. Oncogene. 35:4179–4187. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Sun TT, Wang Y, Cheng H, Xiao HZ, Xiang JJ, Zhang JT, Yu SB, Martin TA, Ye L, Tsang LL, et al: Disrupted interaction between CFTR and AF-6/afadin aggravates malignant phenotypes of colon cancer. Biochim Biophys Acta. 1843:618–628. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Palma AG, Soares Machado M, Lira MC, Rosa F, Rubio MF, Marino G, Kotsias BA and Costas MA: Functional relationship between CFTR and RAC3 expression for maintaining cancer cell stemness in human colorectal cancer. Cell Oncol (Dordr). 44:627–641. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Strubberg AM, Liu J, Walker NM, Stefanski CD, MacLeod RJ, Magness ST and Clarke LL: Cftr modulates Wnt/β-catenin signaling and stem cell proliferation in murine intestine. Cell Mol Gastroenterol Hepatol. 5:253–271. 2017. View Article : Google Scholar | |
|
Liu C, Song C, Li J and Sun Q: CFTR functions as a tumor suppressor and is regulated by DNA methylation in colorectal cancer. Cancer Manag Res. 12:4261–4270. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Paulino C, Neldner Y, Lam AK, Kalienkova V, Brunner JD, Schenck S and Dutzler R: Structural basis for anion conduction in the calcium-activated chloride channel TMEM16A. Elife. 6:e262322017. View Article : Google Scholar : PubMed/NCBI | |
|
Dang S, Feng S, Tien J, Peters CJ, Bulkley D, Lolicato M, Zhao J, Zuberbühler K, Ye W, Qi L, et al: Cryo-EM structures of the TMEM16A calcium-activated chloride channel. Nature. 552:426–429. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Ji Q, Guo S, Wang X, Pang C, Zhan Y, Chen Y and An H: Recent advances in TMEM16A: Structure, function, and disease. J Cell Physiol. 234:7856–7873. 2019. View Article : Google Scholar | |
|
Liu Y, Liu Z and Wang K: The Ca(2+)-activated chloride channel ANO1/TMEM16A: An emerging therapeutic target for epithelium-originated diseases? Acta Pharm Sin B. 11:1412–1433. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Crottès D and Jan LY: The multifaceted role of TMEM16A in cancer. Cell Calcium. 82:1020502019. View Article : Google Scholar : PubMed/NCBI | |
|
Jang Y and Oh U: Anoctamin 1 in secretory epithelia. Cell Calcium. 55:355–361. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Ji Q, Shi S, Guo S, Zhan Y, Zhang H, Chen Y and An H: Activation of TMEM16A by natural product canthaxanthin promotes gastrointestinal contraction. FASEB J. 34:13430–13444. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Stephan AB, Shum EY, Hirsh S, Cygnar KD, Reisert J and Zhao H: ANO2 is the cilial calcium-activated chloride channel that may mediate olfactory amplification. Proc Natl Acad Sci USA. 106:11776–11781. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Huang WC, Xiao S, Huang F, Harfe BD, Jan YN and Jan LY: Calcium-activated chloride channels (CaCCs) regulate action potential and synaptic response in hippocampal neurons. Neuron. 74:179–192. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Ha GE, Lee J, Kwak H, Song K, Kwon J, Jung SY, Hong J, Chang GE, Hwang EM, Shin HS, et al: The Ca(2+)-activated chloride channel anoctamin-2 mediates spike-frequency adaptation and regulates sensory transmission in thalamocortical neurons. Nat Commun. 7:137912016. View Article : Google Scholar : PubMed/NCBI | |
|
Vanoni S, Zeng C, Marella S, Uddin J, Wu D, Arora K, Ptaschinski C, Que J, Noah T, Waggoner L, et al: Identification of anoctamin 1 (ANO1) as a key driver of esophageal epithelial proliferation in eosinophilic esophagitis. J Allergy Clin Immunol. 145:239–254.e2. 2020. View Article : Google Scholar : | |
|
Yu Y, Cao J, Wu W, Zhu Q, Tang Y, Zhu C, Dai J, Li Z, Wang J, Xue L, et al: Genome-wide copy number variation analysis identified ANO1 as a novel oncogene and prognostic biomarker in esophageal squamous cell cancer. Carcinogenesis. 40:1198–1208. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Le SC, Jia Z, Chen J and Yang H: Molecular basis of PIP(2)-dependent regulation of the Ca(2+)-activated chloride channel TMEM16A. Nat Commun. 10:37692019. View Article : Google Scholar : PubMed/NCBI | |
|
Fairfield CJ, Drake TM, Pius R, Bretherick AD, Campbell A, Clark DW, Fallowfield JA, Hayward C, Henderson NC, Iakovliev A, et al: Genome-wide analysis identifies gallstone-susceptibility loci including genes regulating gastrointestinal motility. Hepatology. 75:1081–1094. 2022. View Article : Google Scholar | |
|
Camilleri M, Sandler RS and Peery AF: Etiopathogenetic mechanisms in diverticular disease of the colon. Cell Mol Gastroenterol Hepatol. 9:15–32. 2020. View Article : Google Scholar | |
|
Schafmayer C, Harrison JW, Buch S, Lange C, Reichert MC, Hofer P, Cossais F, Kupcinskas J, von Schönfels W, Schniewind B, et al: Genome-wide association analysis of diverticular disease points towards neuromuscular, connective tissue and epithelial pathomechanisms. Gut. 68:854–865. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zeng X, Pan D, Wu H, Chen H, Yuan W, Zhou J, Shen Z and Chen S: Transcriptional activation of ANO1 promotes gastric cancer progression. Biochem Biophys Res Commun. 512:131–136. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Xie R, Liu L, Lu X and Hu Y: LncRNA OIP5-AS1 facilitates gastric cancer cell growth by targeting the miR-422a/ANO1 axis. Acta Biochim Biophys Sin (Shanghai). 52:430–438. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lu G, Shi W and Zheng H: Inhibition of STAT6/Anoctamin-1 activation suppresses proliferation and invasion of gastric cancer cells. Cancer Biother Radiopharm. 33:3–7. 2018.PubMed/NCBI | |
|
Yan Y, Ding X, Han C, Gao J, Liu Z, Liu Y and Wang K: Involvement of TMEM16A/ANO1 upregulation in the oncogenesis of colorectal cancer. Biochim Biophys Acta Mol Basis Dis. 1868:1663702022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Q, Bai L, Luo S, Wang T, Yang F, Xia J, Wang H, Ma K, Liu M, Wu S, et al: TMEM16A Ca(2+)-activated Cl(-) channel inhibition ameliorates acute pancreatitis via the IP(3) R/Ca(2+)/NFκB/IL-6 signaling pathway. J Adv Res. 23:25–35. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang G, Shu Z, Yu J, Li J, Yi P, Wu B, Deng D, Yan S, Li Y, Ren D, et al: High ANO1 expression is a prognostic factor and correlated with an immunosuppressive tumor microenvironment in pancreatic cancer. Front Immunol. 15:13412092024. View Article : Google Scholar : PubMed/NCBI | |
|
Guo JW, Liu X, Zhang TT, Lin XC, Hong Y, Yu J, Wu QY, Zhang FR, Wu QQ, Shang JY, et al: Hepatocyte TMEM16A deletion retards NAFLD progression by ameliorating hepatic glucose metabolic disorder. Adv Sci (Weinh). 7:19036572020. View Article : Google Scholar : PubMed/NCBI | |
|
Guo J, Song Z, Yu J, Li C, Jin C, Duan W, Liu X, Liu Y, Huang S, Tuo Y, et al: Hepatocyte-specific TMEM16A deficiency alleviates hepatic ischemia/reperfusion injury via suppressing GPX4-mediated ferroptosis. Cell Death Dis. 13:10722022. View Article : Google Scholar : PubMed/NCBI | |
|
Kondo R, Furukawa N, Deguchi A, Kawata N, Suzuki Y, Imaizumi Y and Yamamura H: Downregulation of Ca(2+)-activated Cl(-) channel TMEM16A mediated by angiotensin II in cirrhotic portal hypertensive mice. Front Pharmacol. 13:8313112022. View Article : Google Scholar : PubMed/NCBI | |
|
Deng L, Yang J, Chen H, Ma B, Pan K, Su C, Xu F and Zhang J: Knockdown of TMEM16A suppressed MAPK and inhibited cell proliferation and migration in hepatocellular carcinoma. Onco Targets Ther. 9:325–333. 2016.PubMed/NCBI | |
|
König B and Stauber T: Biophysics and structure-function relationships of LRRC8-formed volume-regulated anion channels. Biophys J. 116:1185–1193. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Sawicka M and Dutzler R: Regulators of cell volume: The structural and functional properties of anion channels of the LRRC8 family. Curr Opin Struct Biol. 74:1023822022. View Article : Google Scholar : PubMed/NCBI | |
|
Okada Y, Sabirov RZ, Sato-Numata K and Numata T: Cell death induction and protection by activation of ubiquitously expressed anion/cation channels. Part 1: Roles of VSOR/VRAC in cell volume regulation, release of double-edged signals and apoptotic/necrotic cell death. Front Cell Dev Biol. 8:6140402021. View Article : Google Scholar : PubMed/NCBI | |
|
Chen L, König B, Liu T, Pervaiz S, Razzaque YS and Stauber T: More than just a pressure relief valve: Physiological roles of volume-regulated LRRC8 anion channels. Biol Chem. 400:1481–1496. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Konishi T, Shiozaki A, Kosuga T, Kudou M, Shoda K, Arita T, Konishi H, Komatsu S, Kubota T, Fujiwara H, et al: LRRC8A expression influences growth of esophageal squamous cell carcinoma. Am J Pathol. 189:1973–1985. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Kurashima K, Shiozaki A, Kudou M, Shimizu H, Arita T, Kosuga T, Konishi H, Komatsu S, Kubota T, Fujiwara H, et al: LRRC8A influences the growth of gastric cancer cells via the p53 signaling pathway. Gastric Cancer. 24:1063–1075. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Xu R, Hu Y, Xie Q, Zhang C, Zhao Y, Zhang H, Shi H, Wang X and Shi C: LRRC8A is a promising prognostic biomarker and therapeutic target for pancreatic adenocarcinoma. Cancers (Basel). 14:55262022. View Article : Google Scholar : PubMed/NCBI | |
|
Lu P, Ding Q, Li X, Ji X, Li L, Fan Y, Xia Y, Tian D and Liu M: SWELL1 promotes cell growth and metastasis of hepatocellular carcinoma in vitro and in vivo. EBioMedicine. 48:100–116. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang H, Deng Z, Zhang D, Li H, Zhang L, Niu J, Zuo W, Fu R, Fan L, Ye JH and She J: High expression of leucine-rich repeat-containing 8A is indicative of a worse outcome of colon cancer patients by enhancing cancer cell growth and metastasis. Oncol Rep. 40:1275–1286. 2018.PubMed/NCBI | |
|
Fujii T, Shimizu T, Yamamoto S, Funayama K, Fujita K, Tabuchi Y, Ikari A, Takeshima H and Sakai H: Crosstalk between Na(+),K(+)-ATPase and a volume-regulated anion channel in membrane microdomains of human cancer cells. Biochim Biophys Acta Mol Basis Dis. 1864:3792–3804. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang H, Jing Z, Liu R, Shada Y, Shria S, Cui S, Ren Y, Wei Y, Li L and Peng S: LRRC8A promotes the initial development of oxaliplatin resistance in colon cancer cells. Heliyon. 9:e168722023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang H, Cui S, Jing Z, Fu G, Liu R, Zhao W, Xu L, Yu L, Bai Y, Lv C, et al: LRRC8A is responsible for exosome biogenesis and volume regulation in colon cancer cells. Biochem J. 480:701–713. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang HY, Shimizu T, Numata T and Okada Y: Role of acid-sensitive outwardly rectifying anion channels in acidosis-induced cell death in human epithelial cells. Pflugers Arch. 454:223–233. 2007. View Article : Google Scholar | |
|
Osei-Owusu J, Kots E, Ruan Z, Mihaljević L, Chen KH, Tamhaney A, Ye X, Lü W, Weinstein H and Qiu Z: Molecular determinants of pH sensing in the proton-activated chloride channel. Proc Natl Acad Sci USA. 119:e22007271192022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao J, Zhu D, Zhang X, Zhang Y, Zhou J and Dong M: TMEM206 promotes the malignancy of colorectal cancer cells by interacting with AKT and extracellular signal-regulated kinase signaling pathways. J Cell Physiol. 234:10888–10898. 2019. View Article : Google Scholar | |
|
Zhang L, Liu SY, Yang X, Wang YQ and Cheng YX: TMEM206 is a potential prognostic marker of hepatocellular carcinoma. Oncol Lett. 20:1742020.PubMed/NCBI | |
|
Huang X, Chen H, Michelsen K, Schneider S and Shaffer PL: Crystal structure of human glycine receptor-α3 bound to antagonist strychnine. Nature. 526:277–280. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Clar DT and Maani CV: Physiology, Ligand Gated Chloride Channel. StatPearls. StatPearls Publishing; Treasure Island, FL: 2024 | |
|
Sanjari Moghaddam H, Zare-Shahabadi A, Rahmani F and Rezaei N: Neurotransmission systems in Parkinson's disease. Rev Neurosci. 28:509–536. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Hsu YT, Chang YG and Chern Y: Insights into GABA(A)ergic system alteration in Huntington's disease. Open Biol. 8:1801652018. View Article : Google Scholar : PubMed/NCBI | |
|
Govindpani K, Calvo-Flores Guzmán B, Vinnakota C, Waldvogel HJ, Faull RL and Kwakowsky A: Towards a better understanding of GABAergic remodeling in Alzheimer's disease. Int J Mol Sci. 18:18132017. View Article : Google Scholar : PubMed/NCBI |