Chloride intracellular channels as novel biomarkers for digestive system tumors (Review)

  • Authors:
    • Hui Wang
    • Jiaxing An
    • Suyu He
    • Chengcheng Liao
    • Juan Wang
    • Biguang Tuo
  • View Affiliations

  • Published online on: July 5, 2021     https://doi.org/10.3892/mmr.2021.12269
  • Article Number: 630
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Digestive system malignant tumors are common tumors, and the traditional treatment methods for these tumors include surgical resection, radiotherapy, chemotherapy, and molecularly targeted drugs. However, diagnosis remains challenging, and the early detection of postoperative recurrence is complicated. Therefore, it is necessary to explore novel biomarkers to facilitate clinical diagnosis and treatment. Accumulating evidence supports the crucial role of chloride channels in the development of multiple types of cancers. Given that chloride channels are widely expressed and involved in cell proliferation, apoptosis and cell cycle, among other processes, they may serve as a promising diagnostic and therapeutic target. Chloride intracellular channels (CLICs) are a class of chloride channels that are upregulated or downregulated in certain types of cancer. Furthermore, in certain cases, during cell cycle progression, the localization and function of the cytosolic form of the transmembrane proteins of CLICs are also altered, which may provide a key target for cancer therapy. The aim of the present review was to focus on CLICs as biomarkers for digestive system tumors.

Introduction

It is estimated that there will be >1.9 million new cases of cancer in the United States in 2021, accompanied by >608,000 deaths (1). Cancer of the digestive system has the second-highest number of new cases and cancer deaths (1). It is widely accepted that the normal function of the digestive system is essential for food digestion, residue excretion, nutrient absorption, and toxic substance discharge. These functions are dependent on the transport of large amounts of water, ions, and nutrients across the epithelium. These physiological processes are achieved via the uneven distribution of ions mediated by ion channels (2), such as absorption of glucose by sodium-glucose cotransporters in the small intestine (3). In addition to controlling the distribution of chloride inside and outside of the cell to maintain water-electrolyte balance, chloride channels also contribute to the regulation of intracellular volume and pH (4). Numerous studies have found that chloride intracellular channels (CLICs) have crucial roles in tumors of the digestive system and should be considered a potential diagnostic and therapeutic target for cancer (59). Therefore, the present review focused on CLICs as novel biomarkers for digestive system tumors.

Structural features of CLICs

In mammals, the CLIC family has seven members, CLIC1-6. Of these, CLIC5 has two alternative splice variants, CLIC5A and CLIC5B. CLICs exist as soluble globular proteins that can form ion channels in organelles and plasma membranes (10). However, to date, only crystal structures of soluble forms of CLICs have been obtained. CLICs comprise an N-terminal thioredoxin-like domain, which contains a mixture of α-helices and β-sheets, and an α-helical C-terminal domain. The N-terminal domain includes a putative transmembrane region (PTM) (4). In the three-dimensional folded structure, there is a similarity between soluble CLICs and ω class glutathione (GSH)-S-transferases (GSTs). CLICs, similar to ω class GST proteins, include a conserved glutaredoxin-like site and a reactive cysteine residue (Cys24 in CLIC1 and Cys35 in CLIC4) in mammals (11), suggesting that the function of CLICs can be regulated in a redox-dependent manner. Furthermore, the CLIC structure includes an elongated cleft (or groove), similar to ω class GSTs, which can bind to glutathione (10). However, CLIC proteins have a very low affinity for glutathione (4). It can therefore be inferred that CLIC proteins use GSH-binding sites to target the CLICs to specific subcellular sites (4). The structure of CLIC1 is presented in Fig. 1. In addition to the N-terminus, C-terminus and PTM, which are common among CLICs, the secondary structure of CLIC1 is also shown. The primary tissues with physiological CLIC protein expression and digestive system tumors presenting abnormal expression of CLIC proteins are presented in Table I (1236).

Table I.

Expression for CLICs and their abnormal expression in digestive system tumors.

Table I.

Expression for CLICs and their abnormal expression in digestive system tumors.

CLIC family memberExpression in normal tissuesType of cancerExpressionAssociation with prognosis(Refs.)
CLIC1Glandular, stomach, small intestine, colon, bile duct, pancreatic duct, airway, the tail of the epididymis, renal, liver, brainESCCUpregulatedPrediction of prognosis of patients with ESCC(1218)
GCUpregulatedPrediction of lymph node metastasis, lymphatic invasion, perineural invasion and poor prognosis
LCUpregulatedPrediction of tumor size, metastasis and pTNM stage and decreased overall survival
Gallbladder cancerUpregulatedPrediction of histological grade, TNM stage, perineural invasion and decreased overall survival
PCUpregulatedPrediction of prognosis of patients with PC
CRCUpregulatedPrediction of prognosis of patients with CRC
CLIC2Liver, heart and skeletal muscle, brain, lung and spleen, stomach and testisESCCUnchangedND(1923)
GCDownregulatedND
LCDownregulatedND
PCUpregulatedND
CLIC3Placental, lung, heart, renal, pancreatic, skeletal muscle (low expression)ESCCDownregulatedND(15,2326)
GCUpregulatedCLIC3 expression is inversely correlated with the depth of tumor invasion, and low CLIC3 expression is associated with prognosis in patients with GC
LCUpregulatedND
PCUpregulatedPrediction of prognosis of patients with PC
CLIC4Skin, brain, liver, testis, renal, lung, skeletal muscleESCCUpregulatedND(15,23,2732)
GCUpregulatedND
LCUpregulatedND
PCUpregulatedPrediction of prognosis of patients with PC
CRCUpregulatedCLIC4 overexpression is a marker of colon cancer stem cells and is associated with poor prognosis
CLIC5CLIC5A: Heart, renal, lung,GCDownregulatedND(15,31,3336)
placental, skeletal, inner ear. CLIC5B: Osteoclast ruffledLCDownregulatedPrediction of prognosis of patients with LC
membrane, heart, skeletal, muscle, renal, inner earPCUpregulatedND
CLIC6Stomach, parietal cell, choroid plexus,GCDownregulatedND(15,34)
salivary duct, lacrimalgland, renal, airway,LCUpregulatedND
and chorioretinal epithelialPCDownregulatedND

[i] CLIC, chloride intracellular channel; ESCC, esophageal squamous cell carcinoma; GC, gastric cancer; LC, liver cancer; PC, pancreatic cancer; CRC, colorectal cancer; ND, not determined.

Functions of CLICs associated with cancer hallmarks

CLIC functions are summarized in Table II (1244). This review focused on CLIC functions that may be associated with hallmarks of cancer. It is well known that tumor cells exhibit a variety of special biological behaviors, including apoptosis evasion, limitless replication potential, sustained angiogenesis, tissue invasion, and metastasis (45). Ion transport exerts crucial roles in tumor development and progression (46,47). The characteristic that distinguishes CLICs from other ion channels is their dimorphic existence. CLICs are located both in the cell membrane and the cytoplasm (10). Of note, CLICs are involved in cancer by regulating ion transport, but the characteristics of their non-ionic transport need to be further elucidated. All CLICs (CLIC1-6) function as ion channels to facilitate chloride flux across cell membranes and, as such, they provide a transcellular route for chloride transport across membranes (21,39,4851). Both the overexpression of p64 in HeLa cells (52) and purified CLIC1 following its expression in Escherichia coli (53) resulted in the appearance of an outwardly rectifying anion channel.

Table II.

Common and unique functions of CLIC family proteins.

Table II.

Common and unique functions of CLIC family proteins.

CLIC family memberUnique functionsCommon functions(Refs.)
CLIC11.Phagosomal acidification1. Chloride channel activity(14,16,17,19,23,2831)
2.Platelet aggregation(CLIC1-CLIC6)
3. Positive regulation of osteoblast2. Endosomal trafficking
differentiation(CLIC3 and CLIC4)
4. Inflammation3. Glutaredoxin-like activity
in vitro (CLIC1, CLIC2 and CLIC4)
4. Regulation of cell cycle
(CLIC1 and CLIC4)
5. Regulation of mitochondrial
membrane potential (CLIC1 and CLIC4)
6. Protein binding (CLIC1, CLIC2, CLIC3 and CLIC5B)
7. Angiogenesis (CLIC1, CLIC4 and CLIC5)
8. Mediating endothelial cell
proliferation (CLIC1 and CLIC4)
9. Migration/metastasis (CLIC1,
CLIC2, CLIC4 and CLIC5)
10. Apoptosis (CLIC4 and CLIC5)
CLIC21. Negative regulation of ryanodine-sensitive (15,17,20,31,32)
calcium-release channel activity
2. Regulation of cardiac muscle
contraction by guiding the release of
sequestered calcium ion
3. Regulation of release of sequestered
calcium ions into cytosol by
sarcoplasmic reticulum
CLIC3Unknown (12,21)
CLIC41. Regulation of TGF-β signaling. (13,18,2427,33,34,35,37,38)
2. Keratinocyte differentiation
3. Establishment or maintenance of
apical/basal cell polarity during mitosis and
cytokinesis
4. Fertilization
5. Vacuolar acidification
CLIC51. CLIC5 is vital for the formation of (22,36,3943)
stereocilia in the inner ear and normal
development of the organ of Corti
2. CLIC5 has a role in female pregnancy
3. CLIC5A has a role in membrane-ERM
interaction in cilia, in the maintenance of
podocyte and glomerular architecture and
in actin-dependent membrane remodeling
4. CLIC5B has a role in actin-dependent
membrane remodeling and in osteoclast
differentiation
CLIC61. D2 dopamine receptor binding (24,44)
2. D3 dopamine receptor binding

[i] CLIC, chloride intracellular channel.

Cl, the most abundant anion in nature, is involved in general biophysical processes, such as the transport of water and osmotic equilibrium (54). Thus far, it is known that for apoptosis to occur, cell volume, DNA fragmentation or division and apoptotic body formation should be tightly controlled (55). Cell volume changes are likely to be partly due to intra and extracellular ionic imbalance. Cell shrinkage is of great importance in the initial step during the apoptotic process and is mediated by the activation of ion channels that release K+, Cl and other organic molecules, followed by the extrusion of water out of the cells by osmotic pressure gradient (56).

Cl could have a critical role in the regulation of cell cycle progression and proliferation (57). The treatment of Jurkat leukemic T-lymphocyte cells with lectins, concanavalin A or phytohemagglutinin resulted in Cl oscillations and increased intracellular chloride concentration [(Cl)i] (58). The lectin-induced [Cl]i increase was blocked by anthracene-9 carboxylate (an inhibitor of Cl channels) or through the removal of extracellular Cl, which inhibited Jurkat cell proliferation (58). Furthermore, [Cl]i may affect the cell cycle through regulating the G1/S cell-cycle checkpoint and p21, which is a cyclin-dependent kinase inhibitor (57). Cell proliferation was reduced in the low [Cl]i conditions following G0/G1 phase arrest in an MKN28 human gastric cancer (GC) cell line. The process involved p21 upregulation caused by low [Cl]i but was independent of p53 (57). In addition, [Cl]i was found to have an essential role in neurite growth in PC12 cells (59). A positive correlation was found between [Cl]i and neurite length (60). For instance, inhibited Na+/K+/2Cl cotransport 1 with bumetanide resulted in a decrease in [Cl]i and neurite outgrowth (60).

In conclusion, [Cl]i has been shown to inhibit proliferation and induce apoptosis. However, Heimlich and Cidlowski (61) reported that both the increased and decreased [Cl]i had the same effect when Jurkat T-cells were exposed to UV-C, which resulted in a significant decrease in [Cl]i and induced apoptosis through the activation of c-Jun N-terminal kinase (JNK). Those processes could be suppressed by the modulation of chloride flux through the reduction of extracellular chloride concentration or in the presence of chloride channel inhibitor disodium 4-acetamido-4′-isothiocyanato-stilben-2,2′-disulfonate, which resulted in an ~20-mM increase in [Cl]I (61). The mechanisms underlying the biphasic effect of [Cl]i need to be further elucidated.

CLICs not only have significant roles in cancer directly through chloride but also through interacting with other proteins and affecting cell signaling. Each CLIC member has unique functions that have been associated with hallmarks of cancer, including cell cycle, apoptosis, angiogenesis, migration and metastasis.

CLICs and the cell cycle

CLIC1 and CLIC4 are involved in cell cycle regulation (16,62). CLIC1 chloride conductance has been found to be altered during the cell cycle, and CLIC1 has been shown to be expressed only on the plasma membrane of Chinese hamster ovary (CHO)-K1 cells in the G2/M phase (16). In addition, the cell cycle has been shown to be arrested in the G2/M stage following treatment with chloride channel blockers indanyloxyacetic acid-94 (IAA-94) or anthracene-9-carboxylic acid (16). At the same stage, CLIC1 is selectively expressed on the plasma membrane (16). Mechanistically, the chloride flow causes the osmotic movement of water to alter the cell volume, which may in turn prevent cell division and/or the dissolution of the nuclear envelope (16). A recent study on the function of CLIC1 in medulloblastoma shed new light on its roles in cell cycle regulation (62). CLIC1-mediated chloride efflux may act synergistically with voltage-gated potassium channel-mediated potassium to decrease cell volume and control cell cycle progression. The abnormal increase in cell size beyond a certain threshold has been shown to lead to a high cytoplasm/nucleus (C/N) ratio, reduce macromolecule biosynthesis and hinder the cell cycle (63). CLIC1, whose activity is increased during mitosis, coordinates with potassium voltage-gated channel subfamily H member 5, a mitosis-specific protein localization on the plasma membrane, to regulate the C/N ratio and prevent cytoplasm dilution (62). Indeed, CLIC1 knockout inhibits the proliferation of tumor cells but does not affect mouse development, thereby increasing the survival of medulloblastoma-bearing mice (62). CLIC4 contributes to cell cycle arrest, possibly due to altering the Cl levels and pH of the nucleus. Furthermore, CLIC4 protein knockdown induces cell cycle arrest in differentiating keratinocytes (34). When keratinocytes undergo growth arrest by differentiation in vitro, the cytoplasmic CLIC4 protein in actively proliferating keratinocytes translocates into the nucleus (34). Furthermore, nuclear Cl is increased by targeting CLIC4 directly to the nucleus through adenoviral transduction in keratinocytes (34).

CLICs and apoptosis

CLIC4 and CLIC5 participate in apoptosis. CLIC4 acts as an apoptotic effector and has a substantial role in p53 and c-Myc-mediated apoptosis (27). p53 overexpression or DNA damage mediates CLIC4 upregulation and induces apoptosis (27). Mechanistically, mitochondrial membrane potential is reduced by CLIC4 overexpression, followed by cytochrome c release into the cytoplasm and the activation of caspases to induce apoptosis (27). CLIC4 downregulation reduces p53-induced, but not Bax-induced apoptosis, indicating that the two pro-apoptotic proteins function independently (27). CLIC4 downregulation enhances autophagy and contributes to mitochondrial and endoplasmic reticulum stress-induced apoptosis under starvation (64). It has been shown that endogenous CLIC4 from the cytoplasm translocates into the nucleus under starvation, as well as after treatment with DNA-damaging agents (etoposide, adriamycin and mitomycin), metabolic inhibitors (cycloheximide and actinomycin D), camptothecin, tumor necrosis factor-α and transforming growth factor-β (Fig. 2) (64,65). CLIC4 nuclear translocation is a response to stress and may contribute to the initiation of apoptosis-related nuclear alterations (65).

CLIC5 is located at the inner mitochondrial membrane and CLIC4 in the outer mitochondrial membrane. CLIC5 has a direct role in the regulation of mitochondrial reactive oxygen species (ROS) generation (66). Mitochondria serve a significant role in lysosomal-mediated cell death (67). The ETS variant transcription factor 6 (ETV6)/RUNX family transcription factor 1 fusion gene results in childhood precursor B-cell acute lymphoblastic leukemia (68). The loss of ETV6 transcriptional repressor induces CLIC5 upregulation, ultimately leading to a decrease in lysosome-mediated apoptosis, indicating that CLIC5 activity facilitates an environment of oxidative stress induced by DNA damage accumulation, thereby contributing to the development of leukemogenesis (40).

CLICs and angiogenesis

The function of angiogenesis involves CLIC1, CLIC4 and CLIC5. The downregulation of CLIC1 reduces endothelial migration, capillary-like network formation, branching morphogenesis and capillary-like sprouting (23). Endothelial cell migration and adhesion depend on an appropriate amount of integrin expression (69). CLIC1 has an essential role in the regulation of the cell surface expression of various integrins in angiogenesis, such as αVβ3, αVβ5 and subunits β1 and α3. In CLIC1−/− mice, CLIC1 knockdown resulted in a mild platelet dysfunction characterized by prolonged bleeding, and P2Y12 receptor signaling-related ADP stimulation led to a reduction in platelet activation (30). The activation of G(12/13) pathways by ADP regulates fibrinogen receptor activation in platelets and dense granule release (70).

The expression of CLIC4 is required at multiple stages of angiogenesis. CLIC4 is necessary for endothelial cell hollowing, a process required for vessel formation during ischemia and embryogenesis (71). CLIC4 promotes endothelial cell proliferation and regulates endothelial morphogenesis (38,72). CLIC4 downregulation was demonstrated to decrease cell proliferation, capillary-like sprouting, lumen formation and capillary network formation, all of which was promoted by CLIC4 upregulation (25). CLIC4−/− mice exhibited defective angiogenesis in vivo (38,71,73). Compared with wild-type mice, CLIC4−/− mice demonstrated abnormal collateral circulation in response to ischemic injury (71,73), and the native cerebral collateral density was reduced, leading to severe infarctions (71), smaller kidneys with fewer glomeruli, less dense peritubular capillary networks (74), and retinal angiogenesis defects (38). Furthermore, CLIC4/CLIC5A-mediated ezrin, radixin, moesin activation is necessary for the maintenance of the glomerular capillary architecture (75).

CLICs and cell cortex-associated migration and metastasis

CLIC1, CLIC2, CLIC4 and CLIC5 interact with the cell cortex (7679). Cell adhesion has a vital role in cancer progression and metastasis. Metastasis is a multi-step process, where cells lose their original tissue contacts across the extracellular matrix (ECM), invade into the surrounding tissue, enter into the blood and/or lymphatic system, extravasate in a distant organ and form new tumors (80). Therefore, tumor cells are significantly influenced by cell-cell and cell-ECM adhesion (80). In order to migrate, cells must interact with their environment through adhesion receptors, such as integrins, and form adhesion complexes that respond to different extracellular cues (81).

CLIC1 and CLIC4 bridge the cortical actin cytoskeleton and the plasma membrane for cytokinesis (76). The downregulation of CLIC4 and CLIC1 result in abnormal blebbing at the polar cortex and regression of the cleavage furrow during late cytokinesis, ultimately forming multinucleated cells (76). CLIC2 is decreased in most endothelial cells in blood vessels of cancer tissue (77). In human umbilical vein endothelial cells (HUVECs), CLIC2 downregulation helps human cancer cells to transmigrate through a HUVEC monolayer (77). CLIC4 is implicated in various actin-based processes, such as integrin trafficking and cell adhesion (78). Mechanistically, CLIC4 regulates the Ras homolog family member A/mouse homolog of diaphanous 2-regulated signaling network to integrate cortical actin assembly and membrane protrusion by binding to profilin-1 (78). In addition, in HeLa and MDA-MB-231 cells, CLIC4 downregulation suppresses cell spreading, cell-matrix adhesion and integrin signaling (82). CLIC4 is recruited to β1 integrin at the plasma membrane and RAB35-positive endosomes by lysophosphatidic acid stimulation. Furthermore, CLIC4 impedes the RAB35-dependent regulation of β1 integrin trafficking by decreasing RAB35 activity (82). In renal glomerular podocyte foot processes, CLIC5A, one of two alternative splicing variants of CLIC5, is a component of the ezrin (EZR)/sodium-hydrogen exchanger regulatory factor 2 (NHERF-2)/podocalyxin cytoskeletal complex (79). Furthermore, at the cell cortex, similar to EZR, CLIC5A may have an essential role in the assembly and/or maintenance of F-actin-based structures (83). CLIC5A is a component of the EZR-NHERF2-podocalyxin complex in glomeruli. In CLIC5-deficient mice, the cytoskeletal association of EZR and NHERF2 was diminished (79). Mechanistically, the interaction of CLIC5A with PI(4,5)P2-generating kinases led to clustered plasma membrane PI(4,5)P2 accumulation, followed by the promotion of EZR activation and actin-dependent cell surface remodeling (79).

CLICs and digestive system tumors

CLICs and esophageal cancer

In esophageal squamous cell carcinoma (ESCC), CLIC1 expression is upregulated in both cell lines (TE2, TE5, TE8, TE9, TE15, KYSE70, KYSE150, KYSE170, and KYSE790), and tissue samples (5). Furthermore, CLIC1 is present in the cytoplasm of cancer cells (5). CLIC1 knockdown inhibited the proliferation of tumor cells, and CLIC1 regulated apoptosis through the Toll-like receptor 2/JNK pathway (5). Furthermore, cell cycle arrest was found to occur in the sub-G1phase following CLIC1 knockdown (5). However, as previously mentioned, CLIC1 is only observed at the plasma membrane of G2/M phase cells, and blockade of CLICK1 by chloride channel blockers IAA-94 and anthracene-9-carboxylic acid resulted in the arrest of CHO-K1 cells in the G2/M phase of the cell cycle (16). Whether this contradictory result was due to the different cell lines or different mechanisms requires further investigation. In addition, patients with strong expression of CLIC1 exhibited a significantly lower 5-year overall survival than those with weak expression of CLIC1 (sample size, 61) (5). In a cohort of 45 patients with ESCC, CLIC3 was found downregulated, CLIC4 was upregulated and CLIC2 unchanged, as detected by quantitative PCR and western blotting (5). The specific functions of CLICs in ESCC, as well as differences in expression, require further study. Squamous cell carcinoma is a common type of esophageal cancer, and CLIC4 has been demonstrated to inhibit the growth of squamous cell carcinoma, and the degree of reduction in CLIC4 coincided with the progression of squamous cell tumors from benign to malignant (5). However, it remains unclear whether CLIC4 is involved in the development and progression of ESCC.

CLICs and GC

In GC, CLIC1 is upregulated and correlates with lymph node metastasis, lymphatic invasion, perineural invasion, and poor patient prognosis (6,84,85). Following CLIC1 knockdown in GC cells SGC-7901, the expression levels of integrin α3, αv and β1 in vivo, as well as the phosphorylation of PI3K/AKT, ERK, and p38, were found to be decreased, while integrin α1 was found to be increased; it was therefore hypothesized that the mechanism of CLIC1 in the progression of GC may be associated with the regulation of integrin family proteins, leading to the sequential regulation of PI3K/AKT, mitogen-activated protein kinase (MAPK)/ERK, and MAPK/p38 pathways (86). CLIC1 upregulation could mediate, at least partly, the ability to enhance invasion and metastasis of GC following the knockdown of proteasome activator subunit 2 (87). As aforementioned, one of the hallmarks of malignancy is angiogenesis, which provides blood supply to the tumor tissue (36). One of the functions of CLICs is their participation in angiogenesis (41,47). Unregulated angiogenesis leads to a constant hypoxia-reoxidation (H-R) state, thus increasing ROS production, providing a substrate for further undifferentiation (88). Similarly, CLIC1 regulates GC cell migration and invasion through the ROS-mediated p38 MAPK signaling pathway (84). This is further confirmed by the fact that blocking CLIC proteins with chloride channel blocker IAA-94 reduces ROS production (89). CLIC1 is closely associated with GC resistance to vincristine, and exosome-mediated transfer of CLIC1 can induce the development of vincristine resistance in vitro, possibly through the upregulation of P-glycoprotein and Bcl-2 (90). Tissue microarray analysis using 107 GC specimens revealed that CLIC3 was inversely correlated with pathological tumor depth, that a lower CLIC3 expression was linked to a worse prognosis, and that CLIC3 functioned as a chloride channel on the plasma membrane of GC cells (91).

CLICs and liver cancer

In hepatitis B virus X protein-positive HepG2 cells, CLIC1 protein accumulation was found to promote hepatocellular carcinoma (HCC) development (92).CLIC1 has been reported to be upregulated in HCC (93), and CLIC1 overexpression in liver tumor tissues was significantly correlated with tumor size, metastasis, and pTNM stage. In addition, CLIC1 overexpression was found to be associated with poor prognosis (7,93). In mouse hepatoma ascites, CLIC1 is expressed in the cytoplasm and plasma membrane in both the Hca-F lymphatic metastasis cell line with a high metastatic potential and the Hca-P lymphatic metastasis cell line with a low metastatic potential. Furthermore, two-dimensional difference-gel electrophoresis revealed that CLIC1 expression was higher in both Hca-F cells and plasma membranes, compared with Hca-P (94). Consistently, another study found that the expression of CLIC1 mRNA and protein in Hca-F cells was higher compared with that in Hca-P cells (2 and 1.6-fold, respectively), and the migration and invasion ability were significantly decreased following CLIC1 downregulation, thus demonstrating that CLIC1 may be a key factor in the development of lymphatic metastasis (95). Furthermore, CLIC1 is upregulated in HCC tissues with portal vein tumor thrombus (96). CLIC1 overexpression was confirmed to decrease maspin expression and increase vascular endothelial growth factor (VEGF), matrix metalloproteinase (MMP) 2, MMP12, and MMP13 expression (96). These results revealed that, in HCC, CLIC1 upregulation was significantly associated with vascular invasion, and that CLIC1 may control the mechanism of HCC invasiveness by targeting maspin (96). CLIC1 downregulation by RNA interference significantly enhanced the expression of tumor metastasis genes annexin A7 and gelsolin in vitro; conversely, annexin A7 and gelsolin downregulation enhanced the expression of CLIC1 in vitro and in vivo (97). These data illustrated that CLIC1 may function by regulating the expression of annexin A7 and gelsolin in the migration and invasion of liver cancer (97). Furthermore, at the transcriptome level, microRNA (miR)-124 directly reduced CLIC1 expression, further inhibiting cell migration, and invasion in HCC cells, but without affecting cell proliferation (98). Alternatively, the levels of CLIC1 can be regulated by miR-122-5P (7).

It is not clear whether CLICs act as ion channels to affect liver cancer. Of note, the chloride channel blocker 4,4′-diisothiocyano-2,2′-stilbenedisulfonic acid (DIDS) can effectively inhibit the proliferation of liver cancer cells (99). DIDS induces G1 arrest by downregulating the protein expression levels of cyclin D1 and cyclin E (99). DIDS reduces the protein expression levels of α-fetoprotein, suggesting that it may be able to improve the prognosis of HCC in patients (99). CLIC1 has a vital role, similar to that of proto-oncogene, that may be targeted in the development of novel tumor treatments with chloride channel blocker IAA-94 (100). However, the high concentration of IAA-94 required for its function, as well as its poor specificity for CLIC1, have affected its use as a CLIC1-specific drug for cancer therapy. Further therapeutic development requires a specific and potent CLIC1 inhibitor. If the structure of soluble CLIC1 were characterized, the rational design of small molecules or peptides for CLIC1 inhibition would provide new avenues for blocking the function of CLIC1 (62). Function inhibition may also be achieved by hyperpolarization using native CLIC1 chloride channels, suggesting a therapeutic modality that does not require gene therapy (101).

CLICs are involved in cytoskeleton formation, while cell deformation is required during tumor invasion and metastasis (102). CLIC2 knockdown in HUVECs allows human cancer cells to migrate through HUVEC monolayers (77). CLIC2 was found to be downregulated in fibrotic and advanced HCC tissues (77). CLIC5 can be used as a biological indicator to predict the prognosis of HCC together with EZR and podocalyxin-like (PODXL) (proteins associated with invasion, migration, and poor prognosis of various types of cancer). It has been found that CLIC5 forms a complex with EZR and PODXL, and that it is required for podocyte structure and function (103). In HCC, EZR, PODXL, and CLIC5 are overexpressed (103). Furthermore, migration and invasion were found to be decreased when the expression of CLIC5 and PODXL was inhibited in Huh7 cells (103).

CLICs and gallbladder cancer (GBC)

In human GBC, CLIC1 expression is upregulated compared with normal tissues, and high CLC1 expression is associated with histological grade, TNM stage, perineural invasion (P<0.05), and decreased overall survival (P<0.001) (104). In fact, CLIC1 expression and histological grade are independent risk factors for overall survival (104). CLIC1 knockdown promoted apoptosis and inhibited proliferation, migration and invasion of GBC cells (105). CLIC1 overexpression promoted GBC-SD18L cell motility and invasion and, conversely, CLIC1 knockdown significantly reduced the in vitro GBC-SD18H cell motility and invasion, indicating that CLIC1 may play an essential role in the metastasis of gallbladder cancer (8). At the transcript level, CLIC1 is a direct target gene of hsa-miR-372 and miR-122 (7,106). In GBC, hsa-miR-372 is downregulated and correlates with the aggressive and progressive behavior of tumors by affecting CLIC1 expression (106). Urothelial cancer associated 1 (UCA1) was found to promote bile duct carcinoma (BDC) cell migration and invasiveness, while miR-122 inhibited their progression (84). CLIC1, as a downstream target gene of miR-122, has the opposite effect. The ERK/MAPK signaling pathway is activated following the upregulation of long non-coding RNA UCA1 (84). UCA1 promoted the metastasis of BDC cells and the activation of the ERK/MAPK pathway by regulating the expression of miR-122 and its downstream gene CLIC1, therefore expanding the options for targeted treatment of cholangiocarcinoma (107). In addition, in GBC, serum carbohydrate antigen 19-9 concentration is positively correlated with the expression levels of troponin T1, slow skeletal type, MMP-9 and CLIC3 (108). Of note, metformin markedly inhibits the proliferation and viability of GBC cells, promotes apoptosis and increases the number of early apoptotic cells (109). Metformin has been further shown to exert growth inhibitory effects by inhibiting p-AKT activity and the Bcl-2 family (85). Of note, either the dysfunction or downregulation of CLIC1 could partially reduce the antitumor effect of metformin, whereas upregulation of CLIC1 could increase drug sensitivity (110).

CLICs and pancreatic cancer

In the pancreas, inhibition of CLIC4 increases β-cell survival, likely due to increased levels of Bcl-2, Bcl-xl, and Bad phosphorylation, and the overexpression of Bcl-2 or Bcl-xl in β-cells increases their resistance to cytokine-induced apoptosis (111). Using in silico modeling to construct an interactome of the CLIC gene family, CLIC1, CLIC3, and CLIC4 have been identified as prognostic markers of overall survival in pancreatic ductal adenocarcinoma (PDAC) compared with healthy controls. Among them, the expressions of CLIC1-CLIC3, CLIC4-CLIC5, and CLIC5-CLIC6 have been found to be positively correlated (112). Similar to these findings, CLIC1 protein expression was significantly increased in tumor samples from patients with resected PDAC compared with normal tissues (67.1% vs. 25.7%, respectively; P<0.001) (113). In addition, CLIC1 overexpression was found to be associated with a higher histological grade, larger tumor size, and worse overall survival [hazard ratio, 5.822; 95% confidence interval (CI), 1.329–15.628; P=0.016) (113). Furthermore, the treatment of pancreatic cancer cell lines with CLIC1-targeting small interfering RNA oligonucleotides significantly reduced cell proliferation, anchorage-independent growth, and cell migration on soft agar (9). CLIC3 drives pancreatic cancer invasiveness by cooperating with RAB25 to regulate α5β1 integrin recycling from late endosomes to the plasma membrane (114). Similarly, CLIC3 predicts lymph node metastasis and poor prognosis in inoperable cases of PDAC (12). CLIC4 and Indian hedgehog have been found to be significantly correlated with tumor grade, lymph node metastasis, tumor invasion, and poor overall survival (115). Of note, HOXA distal transcript (HOTTIP), a long non-coding RNA, is upregulated in PDAC (92), and, by identifying canonical HOTTIP/HOXA13 targets, CLIC5 was found to be crucial for PDAC cell growth and cell invasion (116). However, the oncogenic pathway mediated by HOTTIP is not fully understood.

CLICs and colorectal cancer (CRC)

CLIC1 is overexpressed in CRC tumors (117,118) and is associated with poor prognosis (118). CLIC1 is involved in the metastasis of LOVO colon cancer cells by regulating the ROS/ERK pathway during H-R and regulatory volume decrease (RVD)-mediated chloride channel function (117). Previous studies showed that functionally suppressing CLIC1 using the CLIC1 blocker IAA-94 or CLIC1 knockdown (117,119) inhibited the migration and invasion of colon cancer cells, and the effect was attributed to a decrease in RVD capacity (117). On the other hand, the inhibition of CLIC1 channel activity by IAA94 reduced intracellular ROS production during H-R treatment, resulting in decreased cell migration (96). ROS can regulate CLIC1 translocation and, conversely, CLIC1 chloride current is required for ROS production by NADPH oxidase (120). An increase in CLIC1 current could sustain ROS production, which is necessary for cell cycle progression. This contributes to the development of tumors. It has been reported that CLIC4 expression is a marker of colon cancer stem cells and is associated with poor outcome (121). CLIC4 has also been shown to enhance MMP-9 expression and invasiveness in cancer cell lines evading photodynamic therapy (122). A previous study (121) demonstrated that three proteins, CLIC4, endoplasmic reticulum protein 29 (ERp29), and diablo IAP-binding mitochondrial protein (DIABLO, also known as SMAC), have been identified in metastatic cancer stem-like cells of CRC. The protein expression levels of this three-protein panel (CLIC4, ERp29, and DIABLO) enabled the classification of the validation cohort into risk stratification of colorectal cancer (121).

Conclusion

CLICs have an integral role in the process of tumorigenesis by participating in various physiological processes. In particular, the role of CLIC1 in cancer has been summarized in Fig. 3. The expression levels of CLICs can be used as a diagnostic and prognostic marker for digestive system tumors. However, there is currently a lack of studies based on CLIC knockout mice in digestive system tumors. The understanding of the role of CLICs in health and tumors is incomplete. It is also necessary to clarify whether CLICs function as chloride channels or as proteins under specific disease conditions, and to understand their interactions and exact molecular basis of the complex signaling network activated by CLICs. Subsequently, the potential functional continuity of CLICs between the cytoplasm and membrane needs to be explored. The development of conformation-specific drug inhibitors and CLIC protein activity modulators may herald new and more effective avenues for the treatment of cancer.

Acknowledgments

Not applicable.

Funding

This study was supported by grants from the National Natural Science Foundation of China (grant no. 82073087), the Guizhou Provincial Department of Education Youth Science and Technology Talents Growth Project [grant no. QIAN-JIAO-HE KY ZI (2018)236], and the Zunyi Medical University 2017 New Academic Cultivation and Innovation Exploration Special Project [grant no. Qian-Ke-He-Ping-Tai-Ren-Cai (2017)5733-072].

Availability of data and materials

Not applicable.

Authors' contributions

HW wrote the manuscript. JA and CL participated in information collection, analysis, and organization. SH and JW primarily revised and finalized the manuscript. BT revised the manuscript for clarity and style and critically revised the article for important intellectual content. All authors have read and approved the final manuscript.

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.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2020. CA Cancer J Clin. 70:7–30. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Pácha J: Development of intestinal transport function in mammals. Physiol Rev. 80:1633–1667. 2000. View Article : Google Scholar

3 

Poulsen SB, Fenton RA and Rieg T: Sodium-glucose cotransport. Curr Opin Nephrol Hypertens. 24:463–469. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Argenzio E and Moolenaar WH: Emerging biological roles of Cl-intracellular channel proteins. J Cell Sci. 129:4165–4174. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Kobayashi T, Shiozaki A, Nako Y, Ichikawa D, Kosuga T, Shoda K, Arita T, Konishi H, Komatsu S, Kubota T, et al: Chloride intracellular channel 1 as a switch among tumor behaviors in human esophageal squamous cell carcinoma. Oncotarget. 9:23237–23252. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Ma PF, Chen JQ, Wang Z, Liu JL and Li BP: Function of chloride intracellular channel 1 in gastric cancer cells. World J Gastroenterol. 18:3070–3080. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Jiang X, Liu Y, Wang G, Yao Y, Mei C, Wu X, Ma W and Yuan Y: Up-regulation of CLIC1 activates MYC signaling and forms a positive feedback regulatory loop with MYC in Hepatocellular carcinoma. Am J Cancer Res. 10:2355–2370. 2020.PubMed/NCBI

8 

Wang JW, Peng SY, Li JT, Wang Y, Zhang ZP, Cheng Y, Cheng DQ, Weng WH, Wu XS, Fei XZ, et al: Identification of metastasis-associated proteins involved in gallbladder carcinoma metastasis by proteomic analysis and functional exploration of chloride intracellular channel 1. Cancer Lett. 281:71–81. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Lu J, Dong Q, Zhang B, Wang X, Ye B, Zhang F, Song X, Gao G, Mu J, Wang Z, et al: Chloride intracellular channel 1 (CLIC1) is activated and functions as an oncogene in pancreatic cancer. Med Oncol. 32:6162015. View Article : Google Scholar : PubMed/NCBI

10 

Harrop SJ, DeMaere MZ, Fairlie WD, Reztsova T, Valenzuela SM, Mazzanti M, Tonini R, Qiu MR, Jankova L, Warton K, et al: Crystal structure of a soluble form of the intracellular chloride ion channel CLIC1 (NCC27) at 1.4-A resolution. J Biol Chem. 276:44993–45000. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Littler DR, Harrop SJ, Goodchild SC, Phang JM, Mynott AV, Jiang L, Valenzuela SM, Mazzanti M, Brown LJ, Breit SN and Curmi PM: The enigma of the CLIC proteins: Ion channels, redox proteins, enzymes, scaffolding proteins? FEBS Lett. 584:2093–2101. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Dozynkiewicz MA, Jamieson NB, Macpherson I, Grindlay J, van den Berghe PV, von Thun A, Morton JP, Gourley C, Timpson P, Nixon C, et al: Rab25 and CLIC3 collaborate to promote integrin recycling from late endosomes/lysosomes and drive cancer progression. Dev Cell. 22:131–145. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Chou SY, Hsu KS, Otsu W, Hsu YC, Luo YC, Yeh C, Shehab SS, Chen J, Shieh V, He GA, et al: CLIC4 regulates apical exocytosis and renal tube luminogenesis through retromer- and actin-mediated endocytic trafficking. Nat Commun. 7:104122016. View Article : Google Scholar : PubMed/NCBI

14 

Al Khamici H, Brown LJ, Hossain KR, Hudson AL, Sinclair-Burton AA, Ng JP, Daniel EL, Hare JE, Cornell BA, Curmi PM, et al: Members of the chloride intracellular ion channel protein family demonstrate glutaredoxin-like enzymatic activity. PLoS One. 10:e1156992015. View Article : Google Scholar : PubMed/NCBI

15 

Board PG, Coggan M, Watson S, Gage PW and Dulhunty AF: CLIC-2 modulates cardiac ryanodine receptor Ca2+ release channels. Int J Biochem Cell Biol. 36:1599–1612. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Valenzuela SM, Mazzanti M, Tonini R, Qiu MR, Warton K, Musgrove EA, Campbell TJ and Breit SN: The nuclear chloride ion channel NCC27 is involved in regulation of the cell cycle. J Physiol. 529:541–552. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Yang JY, Jung JY, Cho SW, Choi HJ, Kim SW, Kim SY, Kim HJ, Jang CH, Lee MG, Han J and Shin CS: Chloride intracellular channel 1 regulates osteoblast differentiation. Bone. 45:1175–1185. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Fernández-Salas E, Sagar M, Cheng C, Yuspa SH and Weinberg WC: p53 and tumor necrosis factor alpha regulate the expression of a mitochondrial chloride channel protein. J Biol Chem. 274:36488–36497. 1999. View Article : Google Scholar

19 

Shanks RA, Larocca MC, Berryman M, Edwards JC, Urushidani T, Navarre J and Goldenring JR: AKAP350 at the Golgi apparatus. II. Association of AKAP350 with a novel chloride intracellular channel (CLIC) family member. J Biol Chem. 277:40973–40980. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Fan L, Yu W and Zhu X: Interaction of Sedlin with chloride intracellular channel proteins. FEBS Lett. 540:77–80. 2003. View Article : Google Scholar : PubMed/NCBI

21 

Qian Z, Okuhara D, Abe MK and Rosner MR: Molecular cloning and characterization of a mitogen-activated protein kinase-associated intracellular chloride channel. J Biol Chem. 274:1621–1627. 1999. View Article : Google Scholar : PubMed/NCBI

22 

Edwards JC, Cohen C, Xu W and Schlesinger PH: c-Src control of chloride channel support for osteoclast HCl transport and bone resorption. J Biol Chem. 281:28011–28022. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Tung JJ and Kitajewski J: Chloride intracellular channel 1 functions in endothelial cell growth and migration. J Angiogenes Res. 2:232010. View Article : Google Scholar : PubMed/NCBI

24 

Gaudet P, Livstone MS, Lewis SE and Thomas PD: Phylogenetic-based propagation of functional annotations within the Gene Ontology consortium. Brief Bioinform. 12:449–462. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Tung JJ, Hobert O, Berryman M and Kitajewski J: Chloride intracellular channel 4 is involved in endothelial proliferation and morphogenesis in vitro. Angiogenesis. 12:209–220. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Rønnov-Jessen L, Villadsen R, Edwards JC and Petersen OW: Differential expression of a chloride intracellular channel gene, CLIC4, in transforming growth factor-beta1-mediated conversion of fibroblasts to myofibroblasts. Am J Pathol. 161:471–480. 2002. View Article : Google Scholar

27 

Fernández-Salas E, Suh KS, Speransky VV, Bowers WL, Levy JM, Adams T, Pathak KR, Edwards LE, Hayes DD, Cheng C, et al: mtCLIC/CLIC4, an organellular chloride channel protein, is increased by DNA damage and participates in the apoptotic response to p53. Mol Cell Biol. 22:3610–3620. 2002. View Article : Google Scholar

28 

Salao K, Jiang L, Li H, Tsai VW, Husaini Y, Curmi PM, Brown LJ, Brown DA and Breit SN: CLIC1 regulates dendritic cell antigen processing and presentation by modulating phagosome acidification and proteolysis. Biol Open. 5:620–630. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Qiu MR, Jiang L, Matthaei KI, Schoenwaelder SM, Kuffner T, Mangin P, Joseph JE, Low J, Connor D, Valenzuela SM, et al: Generation and characterization of mice with null mutation of the chloride intracellular channel 1 gene. Genesis. 48:127–136. 2010.PubMed/NCBI

30 

Tang T, Lang X, Xu C, Wang X, Gong T, Yang Y, Cui J, Bai L, Wang J, Jiang W and Zhou R: CLICs-dependent chloride efflux is an essential and proximal upstream event for NLRP3 inflammasome activation. Nat Commun. 8:2022017. View Article : Google Scholar : PubMed/NCBI

31 

Domingo-Fernández R, Coll RC, Kearney J, Breit S and O'Neill LAJ: The intracellular chloride channel proteins CLIC1 and CLIC4 induce IL-1β transcription and activate the NLRP3 inflammasome. J Biol Chem. 292:12077–12087. 2017. View Article : Google Scholar

32 

Dulhunty AF, Pouliquin P, Coggan M, Gage PW and Board PG: A recently identified member of the glutathione transferase structural family modifies cardiac RyR2 substate activity, coupled gating and activation by Ca2+ and ATP. Biochem J. 390:333–343. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Suginta W, Karoulias N, Aitken A and Ashley RH: Chloride intracellular channel protein CLIC4 (p64H1) binds directly to brain dynamin I in a complex containing actin, tubulin and 14-3-3 isoforms. Biochem J. 359:55–64. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Suh KS, Mutoh M, Mutoh T, Li L, Ryscavage A, Crutchley JM, Dumont RA, Cheng C and Yuspa SH: CLIC4 mediates and is required for Ca2+-induced keratinocyte differentiation. J Cell Sci. 120:2631–2640. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Berryman MA and Goldenring JR: CLIC4 is enriched at cell-cell junctions and colocalizes with AKAP350 at the centrosome and midbody of cultured mammalian cells. Cell Motil Cytoskeleton. 56:159–172. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Seco CZ, Oonk AM, Domínguez-Ruiz M, Draaisma JM, Gandía M, Oostrik J, Neveling K, Kunst HP, Hoefsloot LH, del Castillo I, et al: Progressive hearing loss and vestibular dysfunction caused by a homozygous nonsense mutation in CLIC5. Eur J Hum Genet. 23:189–194. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Ling CK, Santos LL, Zhou W and Dimitriadis E: Chloride intracellular channel 4 is dysregulated in endometrium of women with infertility and alters receptivity. Biochem Biophys Res Commun. 531:490–496. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Ulmasov B, Bruno J, Gordon N, Hartnett ME and Edwards JC: Chloride intracellular channel protein-4 functions in angiogenesis by supporting acidification of vacuoles along the intracellular tubulogenic pathway. Am J Pathol. 174:1084–1096. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Berryman M and Bretscher A: Identification of a novel member of the chloride intracellular channel gene family (CLIC5) that associates with the actin cytoskeleton of placental microvilli. Mol Biol Cell. 11:1509–1521. 2000. View Article : Google Scholar : PubMed/NCBI

40 

Pierchala BA, Muñoz MR and Tsui CC: Proteomic analysis of the slit diaphragm complex: CLIC5 is a protein critical for podocyte morphology and function. Kidney Int. 78:868–882. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Ponnalagu D, Rao SG, Farber J, Xin W, Hussain AT, Shah K, Tanda S, Berryman MA, Edwards JC and Singh H: Data supporting characterization of CLIC1, CLIC4, CLIC5 and DmCLIC antibodies and localization of CLICs in endoplasmic reticulum of cardiomyocytes. Data Brief. 7:1038–1044. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Tavasoli M, Li L, Al-Momany A, Zhu LF, Adam BA, Wang Z and Ballermann BJ: The chloride intracellular channel 5A stimulates podocyte Rac1, protecting against hypertension-induced glomerular injury. Kidney Int. 89:833–847. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Redhead C, Sullivan SK, Koseki C, Fujiwara K and Edwards JC: Subcellular distribution and targeting of the intracellular chloride channel p64. Mol Biol Cell. 8:691–704. 1997. View Article : Google Scholar : PubMed/NCBI

44 

Griffon N, Jeanneteau F, Prieur F, Diaz J and Sokoloff P: CLIC6, a member of the intracellular chloride channel family, interacts with dopamine D(2)-like receptors. Brain Res Mol Brain Res. 117:47–57. 2003. View Article : Google Scholar : PubMed/NCBI

45 

Rojo de la Vega M, Chapman E and Zhang DD: NRF2 and the hallmarks of cancer. Cancer Cell. 34:21–43. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Prevarskaya N, Skryma R and Shuba Y: Ion channels in cancer: Are cancer hallmarks oncochannelopathies? Physiol Rev. 98:559–621. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Leanza L, Romio M, Becker KA, Azzolini M, Trentin L, Managò A, Venturini E, Zaccagnino A, Mattarei A, Carraretto L, et al: Direct pharmacological targeting of a mitochondrial ion channel selectively kills tumor cells in vivo. Cancer Cell. 31:516–531.e10. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Valenzuela SM, Martin DK, Por SB, Robbins JM, Warton K, Bootcov MR, Schofield PR, Campbell TJ and Breit SN: Molecular cloning and expression of a chloride ion channel of cell nuclei. J Biol Chem. 272:12575–12582. 1997. View Article : Google Scholar : PubMed/NCBI

49 

Cromer BA, Gorman MA, Hansen G, Adams JJ, Coggan M, Littler DR, Brown LJ, Mazzanti M, Breit SN, Curmi PM, et al: Structure of the Janus protein human CLIC2. J Mol Biol. 374:719–731. 2007. View Article : Google Scholar : PubMed/NCBI

50 

Edwards JC: A novel p64-related Cl-channel: Subcellular distribution and nephron segment-specific expression. Am J Physiol. 276:F398–F408. 1999.PubMed/NCBI

51 

Sachs G, Shin JM, Vagin O, Lambrecht N, Yakubov I and Munson K: The gastric H,K ATPase as a drug target: Past, present, and future. J Clin Gastroenterol. 41 (Suppl 2):S226–S242. 2007. View Article : Google Scholar : PubMed/NCBI

52 

Edwards JC, Tulk B and Schlesinger PH: Functional expression of p64, an intracellular chloride channel protein. J Membr Biol. 163:119–127. 1998. View Article : Google Scholar : PubMed/NCBI

53 

Tulk BM, Schlesinger PH, Kapadia SA and Edwards JC: CLIC-1 functions as a chloride channel when expressed and purified from bacteria. J Biol Chem. 275:26986–26993. 2000. View Article : Google Scholar : PubMed/NCBI

54 

Valdivieso ÁG and Santa-Coloma TA: The chloride anion as a signalling effector. Biol Rev Camb Philos Soc. 94:1839–1856. 2019. View Article : Google Scholar : PubMed/NCBI

55 

Bortner CD, Sifre MI and Cidlowski JA: Cationic gradient reversal and cytoskeleton-independent volume regulatory pathways define an early stage of apoptosis. J Biol Chem. 283:7219–7229. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Kunzelmann K: Ion channels in regulated cell death. Cell Mol Life Sci. 73:2387–2403. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Miyazaki H, Shiozaki A, Niisato N, Ohsawa R, Itoi H, Ueda Y, Otsuji E, Yamagishi H, Iwasaki Y, Nakano T, et al: Chloride ions control the G1/S cell-cycle checkpoint by regulating the expression of p21 through a p53-independent pathway in human gastric cancer cells. Biochem Biophys Res Commun. 366:506–512. 2008. View Article : Google Scholar : PubMed/NCBI

58 

Lai ZF, Chen YZ and Nishi K: Modulation of intracellular Cl-homeostasis by lectin-stimulation in Jurkat T lymphocytes. Eur J Pharmacol. 482:1–8. 2003. View Article : Google Scholar : PubMed/NCBI

59 

Hiraoka K, Miyazaki H, Niisato N, Iwasaki Y, Kawauchi A, Miki T and Marunaka Y: Chloride ion modulates cell proliferation of human androgen-independent prostatic cancer cell. Cell Physiol Biochem. 25:379–388. 2010. View Article : Google Scholar : PubMed/NCBI

60 

Nakajima KI and Marunaka Y: Intracellular chloride ion concentration in differentiating neuronal cell and its role in growing neurite. Biochem Biophys Res Commun. 479:338–342. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Heimlich G and Cidlowski JA: Selective role of intracellular chloride in the regulation of the intrinsic but not extrinsic pathway of apoptosis in Jurkat T-cells. J Biol Chem. 281:2232–2241. 2006. View Article : Google Scholar : PubMed/NCBI

62 

Francisco MA, Wanggou S, Fan JJ, Dong W, Chen X, Momin A, Abeysundara N, Min HK, Chan J, McAdam R, et al: Chloride intracellular channel 1 cooperates with potassium channel EAG2 to promote medulloblastoma growth. J Exp Med. 217:e201909712020. View Article : Google Scholar : PubMed/NCBI

63 

Neurohr GE, Terry RL, Lengefeld J, Bonney M, Brittingham GP, Moretto F, Miettinen TP, Vaites LP, Soares LM, Paulo JA, et al: Excessive cell growth causes cytoplasm dilution and contributes to senescence. Cell. 176:1083–1097.e18. 2019. View Article : Google Scholar : PubMed/NCBI

64 

Zhong J, Kong X, Zhang H, Yu C, Xu Y, Kang J, Yu H, Yi H, Yang X and Sun L: Inhibition of CLIC4 enhances autophagy and triggers mitochondrial and ER stress-induced apoptosis in human glioma U251 cells under starvation. PLoS One. 7:e393782012. View Article : Google Scholar : PubMed/NCBI

65 

Suh KS, Mutoh M, Nagashima K, Fernandez-Salas E, Edwards LE, Hayes DD, Crutchley JM, Marin KG, Dumont RA, Levy JM, et al: The organellular chloride channel protein CLIC4/mtCLIC translocates to the nucleus in response to cellular stress and accelerates apoptosis. J Biol Chem. 279:4632–4641. 2004. View Article : Google Scholar : PubMed/NCBI

66 

Ponnalagu D, Gururaja Rao S, Farber J, Xin W, Hussain AT, Shah K, Tanda S, Berryman M, Edwards JC and Singh H: Molecular identity of cardiac mitochondrial chloride intracellular channel proteins. Mitochondrion. 27:6–14. 2016. View Article : Google Scholar : PubMed/NCBI

67 

Kim R, Emi M and Tanabe K: Role of mitochondria as the gardens of cell death. Cancer Chemother Pharmacol. 57:545–553. 2006. View Article : Google Scholar : PubMed/NCBI

68 

Neveu B, Spinella JF, Richer C, Lagacé K, Cassart P, Lajoie M, Jananji S, Drouin S, Healy J, Hickson GR, et al: CLIC5: A novel ETV6 target gene in childhood acute lymphoblastic leukemia. Haematologica. 101:1534–1543. 2016. View Article : Google Scholar : PubMed/NCBI

69 

Sales A, Ende K, Diemer J, Kyvik AR, Veciana J, Ratera I, Kemkemer R, Spatz JP and Guasch J: Cell Type-dependent integrin distribution in adhesion and migration responses on protein-coated microgrooved substrates. ACS Omega. 4:1791–1800. 2019. View Article : Google Scholar

70 

Jin J, Mao Y, Thomas D, Kim S, Daniel JL and Kunapuli SP: RhoA downstream of G(q) and G(12/13) pathways regulates protease-activated receptor-mediated dense granule release in platelets. Biochem Pharmacol. 77:835–844. 2009. View Article : Google Scholar : PubMed/NCBI

71 

Chalothorn D, Zhang H, Smith JE, Edwards JC and Faber JE: Chloride intracellular channel-4 is a determinant of native collateral formation in skeletal muscle and brain. Circ Res. 105:89–98. 2009. View Article : Google Scholar : PubMed/NCBI

72 

Bohman S, Matsumoto T, Suh K, Dimberg A, Jakobsson L, Yuspa S and Claesson-Welsh L: Proteomic analysis of vascular endothelial growth factor-induced endothelial cell differentiation reveals a role for chloride intracellular channel 4 (CLIC4) in tubular morphogenesis. J Biol Chem. 280:42397–42404. 2005. View Article : Google Scholar : PubMed/NCBI

73 

Lucitti JL, Tarte NJ and Faber JE: Chloride intracellular channel 4 is required for maturation of the cerebral collateral circulation. Am J Physiol Heart Circ Physiol. 309:H1141–H1150. 2015. View Article : Google Scholar : PubMed/NCBI

74 

Edwards JC, Bruno J, Key P and Cheng YW: Absence of chloride intracellular channel 4 (CLIC4) predisposes to acute kidney injury but has minimal impact on recovery. BMC Nephrol. 15:542014. View Article : Google Scholar : PubMed/NCBI

75 

Tavasoli M, Al-Momany A, Wang X, Li L, Edwards JC and Ballermann BJ: Both CLIC4 and CLIC5A activate ERM proteins in glomerular endothelium. Am J Physiol Renal Physiol. 311:F945–F957. 2016. View Article : Google Scholar : PubMed/NCBI

76 

Uretmen Kagiali ZC, Saner N, Akdag M, Sanal E, Degirmenci BS, Mollaoglu G and Ozlu N: CLIC4 and CLIC1 bridge plasma membrane and cortical actin network for a successful cytokinesis. Life Sci Alliance. 3:e2019005582019. View Article : Google Scholar : PubMed/NCBI

77 

Ueno Y, Ozaki S, Umakoshi A, Yano H, Choudhury ME, Abe N, Sumida Y, Kuwabara J, Uchida R, Islam A, et al: Chloride intracellular channel protein 2 in cancer and non-cancer human tissues: Relationship with tight junctions. Tissue Barriers. 7:15937752019. View Article : Google Scholar : PubMed/NCBI

78 

Argenzio E, Klarenbeek J, Kedziora KM, Nahidiazar L, Isogai T, Perrakis A, Jalink K, Moolenaar WH and Innocenti M: Profilin binding couples chloride intracellular channel protein CLIC4 to RhoA-mDia2 signaling and filopodium formation. J Biol Chem. 293:19161–19176. 2018. View Article : Google Scholar : PubMed/NCBI

79 

Al-Momany A, Li L, Alexander RT and Ballermann BJ: Clustered PI(4,5)P2 accumulation and ezrin phosphorylation in response to CLIC5A. J Cell Sci. 127:5164–5178. 2014.PubMed/NCBI

80 

Oh ES, Seiki M, Gotte M and Chung J: Cell adhesion in cancer. Int J Cell Biol. 2012:9656182012. View Article : Google Scholar : PubMed/NCBI

81 

Conway JRW and Jacquemet G: Cell matrix adhesion in cell migration. Essays Biochem. 63:535–551. 2019. View Article : Google Scholar : PubMed/NCBI

82 

Argenzio E, Margadant C, Leyton-Puig D, Janssen H, Jalink K, Sonnenberg A and Moolenaar WH: CLIC4 regulates cell adhesion and β1 integrin trafficking. J Cell Sci. 127:5189–5203. 2014.PubMed/NCBI

83 

Berryman M, Bruno J, Price J and Edwards JC: CLIC-5A functions as a chloride channel in vitro and associates with the cortical actin cytoskeleton in vitro and in vivo. J Biol Chem. 279:34794–34801. 2004. View Article : Google Scholar : PubMed/NCBI

84 

Zhao W, Lu M and Zhang Q: Chloride intracellular channel 1 regulates migration and invasion in gastric cancer by triggering the ROS-mediated p38 MAPK signaling pathway. Mol Med Rep. 12:8041–8047. 2015. View Article : Google Scholar : PubMed/NCBI

85 

Chen CD, Wang CS, Huang YH, Chien KY, Liang Y, Chen WJ and Lin KH: Overexpression of CLIC1 in human gastric carcinoma and its clinicopathological significance. Proteomics. 7:155–167. 2007. View Article : Google Scholar : PubMed/NCBI

86 

Li BP, Mao YT, Wang Z, Chen YY, Wang Y, Zhai CY, Shi B, Liu SY, Liu JL and Chen JQ: CLIC1 promotes the progression of gastric cancer by regulating the MAPK/AKT pathways. Cell Physiol Biochem. 46:907–924. 2018. View Article : Google Scholar : PubMed/NCBI

87 

Zheng DL, Huang QL, Zhou F, Huang QJ, Lin JY and Lin X: PA28β regulates cell invasion of gastric cancer via modulating the expression of chloride intracellular channel 1. J Cell Biochem. 113:1537–1546. 2012.PubMed/NCBI

88 

Krock BL, Skuli N and Simon MC: Hypoxia-induced angiogenesis: Good and evil. Genes Cancer. 2:1117–1133. 2011. View Article : Google Scholar : PubMed/NCBI

89 

Ponnalagu D and Singh H: Anion channels of mitochondria. Handb Exp Pharmacol. 240:71–101. 2017. View Article : Google Scholar : PubMed/NCBI

90 

Zhao K, Wang Z, Li X, Liu JL, Tian L and Chen JQ: Exosome-mediated transfer of CLIC1 contributes to the vincristine-resistance in gastric cancer. Mol Cell Biochem. 462:97–105. 2019. View Article : Google Scholar : PubMed/NCBI

91 

Kawai S and Fujii T, Shimizu T, Sukegawa K, Hashimoto I, Okumura T, Nagata T, Sakai H and Fujii T: Pathophysiological properties of CLIC3 chloride channel in human gastric cancer cells. J Physiol Sci. 70:152020. View Article : Google Scholar : PubMed/NCBI

92 

Li WH, Miao XH, Qi ZT, Ni W, Zhu SY and Fang F: Proteomic analysis of differently expressed proteins in human hepatocellular carcinoma cell lines HepG2 with transfecting hepatitis B virus X gene. Chin Med J (Engl). 122:15–23. 2009.PubMed/NCBI

93 

Zhang S, Wang XM, Yin ZY, Zhao WX, Zhou JY, Zhao BX and Liu PG: Chloride intracellular channel 1 is overexpression in hepatic tumor and correlates with a poor prognosis. APMIS. 121:1047–1053. 2013. View Article : Google Scholar : PubMed/NCBI

94 

Song MY, Tang JW, Sun MZ, Liu SQ and Wang B: Localization and expression of CLIC1 in hepatocarcinoma ascites cell lines with high or low potentials of lymphatic spread. Zhonghua Bing Li Xue Za Zhi. 39:463–466. 2010.(In Chinese). PubMed/NCBI

95 

Li RK, Zhang J, Zhang YH, Li ML, Wang M and Tang JW: Chloride intracellular channel 1 is an important factor in the lymphatic metastasis of hepatocarcinoma. Biomed Pharmacother. 66:167–172. 2012. View Article : Google Scholar : PubMed/NCBI

96 

Wei X, Li J, Xie H, Wang H, Wang J, Zhang X, Zhuang R, Lu D, Ling Q, Zhou L, et al: Chloride intracellular channel 1 participates in migration and invasion of hepatocellular carcinoma by targeting maspin. J Gastroenterol Hepatol. 30:208–216. 2015. View Article : Google Scholar : PubMed/NCBI

97 

Zhang J, Li M, Song M, Chen W, Mao J, Song L, Wei Y, Huang Y and Tang J: Clic1 plays a role in mouse hepatocarcinoma via modulating Annexin A7 and Gelsolin in vitro and in vivo. Biomed Pharmacother. 69:416–419. 2015. View Article : Google Scholar : PubMed/NCBI

98 

Yue X, Cui Y, You Q, Lu Y and Zhang J: MicroRNA-124 negatively regulates chloride intracellular channel 1 to suppress the migration and invasion of liver cancer cells. Oncol Rep. 42:1380–1390. 2019.PubMed/NCBI

99 

Wang R, Kang B, Hu R, Huang Y, Qin Z, Du J and Lin X: ClC-3 chloride channel protein induces G1 arrest in hepatocellular carcinoma Hep3B cells. Oncol Rep. 40:472–478. 2018.PubMed/NCBI

100 

Li X and Weinman SA: Chloride channels and hepatocellular function: Prospects for molecular identification. Annu Rev Physiol. 64:609–633. 2002. View Article : Google Scholar : PubMed/NCBI

101 

Chernet BT and Levin M: Transmembrane voltage potential of somatic cells controls oncogene-mediated tumorigenesis at long-range. Oncotarget. 5:3287–3306. 2014. View Article : Google Scholar : PubMed/NCBI

102 

Liu Z, Lee SJ, Park S, Konstantopoulos K, Glunde K, Chen Y and Barman I: Cancer cells display increased migration and deformability in pace with metastatic progression. FASEB J. 34:9307–9315. 2020. View Article : Google Scholar : PubMed/NCBI

103 

Flores-Téllez TN, Lopez TV, Vásquez Garzón VR and Villa-Treviño S: Co-Expression of Ezrin-CLIC5-podocalyxin is associated with migration and invasiveness in hepatocellular carcinoma. PLoS One. 10:e01316052015. View Article : Google Scholar

104 

Ding Q, Li M, Wu X, Zhang L, Wu W, Ding Q, Weng H, Wang X and Liu Y: CLIC1 overexpression is associated with poor prognosis in gallbladder cancer. Tumour Biol. 36:193–198. 2015. View Article : Google Scholar : PubMed/NCBI

105 

He YM, Zhang ZL, Liu QY, Xiao YS, Wei L, Xi C and Nan X: Effect of CLIC1 gene silencing on proliferation, migration, invasion and apoptosis of human gallbladder cancer cells. J Cell Mol Med. 22:2569–2579. 2018. View Article : Google Scholar : PubMed/NCBI

106 

Zhou N, Cheng W, Peng C, Liu Y and Jiang B: Decreased expression of hsa-miR-372 predicts poor prognosis in patients with gallbladder cancer by affecting chloride intracellular channel 1. Mol Med Rep. 16:7848–7854. 2017. View Article : Google Scholar : PubMed/NCBI

107 

Kong L, Wu Q, Zhao L, Ye J, Li N and Yang H: Upregulated lncRNA-UCA1 contributes to metastasis of bile duct carcinoma through regulation of miR-122/CLIC1 and activation of the ERK/MAPK signaling pathway. Cell Cycle. 18:1212–1228. 2019. View Article : Google Scholar : PubMed/NCBI

108 

Guo Z, Neilson LJ, Zhong H, Murray PS, Zanivan S and Zaidel-Bar R: E-cadherin interactome complexity and robustness resolved by quantitative proteomics. Sci Signal. 7:rs72014. View Article : Google Scholar : PubMed/NCBI

109 

Gu X, Li B, Jiang M, Fang M, Ji J, Wang A, Wang M, Jiang X and Gao C: RNA sequencing reveals differentially expressed genes as potential diagnostic and prognostic indicators of gallbladder carcinoma. Oncotarget. 6:20661–20671. 2015. View Article : Google Scholar : PubMed/NCBI

110 

Liu Y, Wang Z, Li M, Ye Y, Xu Y, Zhang Y, Yuan R, Jin Y, Hao Y, Jiang L, et al: Chloride intracellular channel 1 regulates the antineoplastic effects of metformin in gallbladder cancer cells. Cancer Sci. 108:1240–1252. 2017. View Article : Google Scholar : PubMed/NCBI

111 

Patel D, Ythier D, Brozzi F, Eizirik DL and Thorens B: Clic4, a novel protein that sensitizes β-cells to apoptosis. Mol Metab. 4:253–264. 2015. View Article : Google Scholar : PubMed/NCBI

112 

Magouliotis DE, Sakellaridis N, Dimas K, Tasiopoulou VS, Svokos KA, Svokos AA and Zacharoulis D: In silico Transcriptomic analysis of the chloride intracellular channels (CLIC) interactome identifies a molecular panel of seven prognostic markers in patients with pancreatic ductal adenocarcinoma. Curr Genomics. 21:119–127. 2020. View Article : Google Scholar : PubMed/NCBI

113 

Jia N, Dong S, Zhao G, Gao H, Li X and Zhang H: CLIC1 overexpression is associated with poor prognosis in pancreatic ductal adenocarcinomas. J Cancer Res Ther. 12:892–896. 2016. View Article : Google Scholar : PubMed/NCBI

114 

Macpherson IR, Rainero E, Mitchell LE, van den Berghe PV, Speirs C, Dozynkiewicz MA, Chaudhary S, Kalna G, Edwards J, Timpson P, et al: CLIC3 controls recycling of late endosomal MT1-MMP and dictates invasion and metastasis in breast cancer. J Cell Sci. 127:3893–3901. 2014.PubMed/NCBI

115 

Zou Q, Yang Z, Li D, Liu Z and Yuan Y: Association of chloride intracellular channel 4 and Indian hedgehog proteins with survival of patients with pancreatic ductal adenocarcinoma. Int J Exp Pathol. 97:422–429. 2016. View Article : Google Scholar : PubMed/NCBI

116 

Wong CH, Li CH, He Q, Chan SL, Tong JH, To KF, Lin LZ and Chen Y: Ectopic HOTTIP expression induces noncanonical transactivation pathways to promote growth and invasiveness in pancreatic ductal adenocarcinoma. Cancer Lett. 477:1–9. 2020. View Article : Google Scholar : PubMed/NCBI

117 

Wang P, Zhang C, Yu P, Tang B, Liu T, Cui H and Xu J: Regulation of colon cancer cell migration and invasion by CLIC1-mediated RVD. Mol Cell Biochem. 365:313–321. 2012. View Article : Google Scholar : PubMed/NCBI

118 

Petrova DT, Asif AR, Armstrong VW, Dimova I, Toshev S, Yaramov N, Oellerich M and Toncheva D: Expression of chloride intracellular channel protein 1 (CLIC1) and tumor protein D52 (TPD52) as potential biomarkers for colorectal cancer. Clin Biochem. 41:1224–1236. 2008. View Article : Google Scholar : PubMed/NCBI

119 

Wang P, Zeng Y, Liu T, Zhang C, Yu PW, Hao YX, Luo HX and Liu G: Chloride intracellular channel 1 regulates colon cancer cell migration and invasion through ROS/ERK pathway. World J Gastroenterol. 20:2071–2078. 2014. View Article : Google Scholar : PubMed/NCBI

120 

Milton RH, Abeti R, Averaimo S, DeBiasi S, Vitellaro L, Jiang L, Curmi PM, Breit SN, Duchen MR and Mazzanti M: CLIC1 function is required for beta-amyloid-induced generation of reactive oxygen species by microglia. J Neurosci. 28:11488–11499. 2008. View Article : Google Scholar : PubMed/NCBI

121 

Deng YJ, Tang N, Liu C, Zhang JY, An SL, Peng YL, Ma LL, Li GQ, Jiang Q, Hu CT, et al: CLIC4, ERp29, and Smac/DIABLO derived from metastatic cancer stem-like cells stratify prognostic risks of colorectal cancer. Clin Cancer Res. 20:3809–3817. 2014. View Article : Google Scholar : PubMed/NCBI

122 

Chiang PC, Chou RH, Chien HF, Tsai T and Chen CT: Chloride intracellular channel 4 involves in the reduced invasiveness of cancer cells treated by photodynamic therapy. Lasers Surg Med. 45:38–47. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2021
Volume 24 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang H, An J, He S, Liao C, Wang J and Tuo B: Chloride intracellular channels as novel biomarkers for digestive system tumors (Review). Mol Med Rep 24: 630, 2021
APA
Wang, H., An, J., He, S., Liao, C., Wang, J., & Tuo, B. (2021). Chloride intracellular channels as novel biomarkers for digestive system tumors (Review). Molecular Medicine Reports, 24, 630. https://doi.org/10.3892/mmr.2021.12269
MLA
Wang, H., An, J., He, S., Liao, C., Wang, J., Tuo, B."Chloride intracellular channels as novel biomarkers for digestive system tumors (Review)". Molecular Medicine Reports 24.3 (2021): 630.
Chicago
Wang, H., An, J., He, S., Liao, C., Wang, J., Tuo, B."Chloride intracellular channels as novel biomarkers for digestive system tumors (Review)". Molecular Medicine Reports 24, no. 3 (2021): 630. https://doi.org/10.3892/mmr.2021.12269