Next Article in Journal
Second-Harmonic Generation Imaging Reveals Changes in Breast Tumor Collagen Induced by Neoadjuvant Chemotherapy
Previous Article in Journal
HPRT1 Promotes Chemoresistance in Oral Squamous Cell Carcinoma via Activating MMP1/PI3K/Akt Signaling Pathway
Previous Article in Special Issue
The Release of Inflammatory Mediators from Acid-Stimulated Mesenchymal Stromal Cells Favours Tumour Invasiveness and Metastasis in Osteosarcoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Chloride Channels and Transporters: Roles beyond Classical Cellular Homeostatic pH or Ion Balance in Cancers

by
Hyeong Jae Kim
1,
Peter Chang-Whan Lee
2,* and
Jeong Hee Hong
1,*
1
Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 155 Getbeolro, Yeonsu-gu, Incheon 21999, Korea
2
Department of Biomedical Sciences, Lung Cancer Research Center, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
*
Authors to whom correspondence should be addressed.
Cancers 2022, 14(4), 856; https://doi.org/10.3390/cancers14040856
Submission received: 31 December 2021 / Revised: 28 January 2022 / Accepted: 6 February 2022 / Published: 9 February 2022
(This article belongs to the Special Issue Tumor Acidosis)

Abstract

:

Simple Summary

Roles of chloride-associated transporters have been raised in various cancers. Although complicated ion movements, crosstalk among channels/transporters through homeostatic electric regulation, difficulties with experimental implementation such as activity measurement of intracellular location were disturbed to verify the precise modulation of channels/transporters, recently defined cancerous function and communication with tumor microenvironment of chloride channels/transporters should be highlighted beyond classical homeostatic ion balance. Chloride-associated transporters as membrane-associated components of chloride movement, regulations of transmembrane member 16A, calcium-activated chloride channel regulators, transmembrane member 206, chloride intracellular channels, voltage-gated chloride channels, cystic fibrosis transmembrane conductance regulator, voltage-dependent anion channel, volume-regulated anion channel, and chloride-bicarbonate exchangers are discussed.

Abstract

The canonical roles of chloride channels and chloride-associated transporters have been physiologically determined; these roles include the maintenance of membrane potential, pH balance, and volume regulation and subsequent cellular functions such as autophagy and cellular proliferative processes. However, chloride channels/transporters also play other roles, beyond these classical function, in cancerous tissues and under specific conditions. Here, we focused on the chloride channel-associated cancers and present recent advances in understanding the environments of various types of cancer caused by the participation of many chloride channel or transporters families and discuss the challenges and potential targets for cancer treatment. The modulation of chloride channels/transporters might promote new aspect of cancer treatment strategies.

1. Chloride Transport

Electrolytes, such as charged anion chloride, drive cellular electrical shifting energy, which mediates various cellular processes. Intracellular chloride ions are abundant (5–40 mM) as are sodium ions [1]. The movement of chloride is considered to regulate cellular membrane potential, cellular volume, and electrostatic compensation as well as maintain the pH of cellular or intra-organelles such as lysosomes. In addition to its classical roles, chloride channels participate in modulation of the cellular fate and motility of cancer cells. Therefore, the finding that these channels function in malignant conditions beyond simply transporting chloride is meaningful. This review summarizes the prevalence and roles of several families of chloride channels/transporters associated with malignant environments and might facilitate a better understanding of cancer and aid in the identification of potential targeted anticancer agents with scope of chloride channel/transporter-based tumorigenesis.

2. Membrane-Associated Components of Chloride Movement

2.1. Transmembrane Member 16A

Transmembrane member (TMEM) 16A (calcium-activated chloride channel; anoctamin 1, ANO1) transports chloride and bicarbonate and plays a role in the proliferation and development of malignant cell types. The expression of TMEM16A has been identified in a broad range of cancers such as non-small cell lung cancer (NSCLC) [2], pancreatic cancer [3], prostate cancer [4], breast cancer [5], colorectal carcinoma [6], gastric cancer [7], glioma [8], glioblastoma [9], esophageal cancer [10], lung cancer [11], hepatocellular carcinoma (HCC) [12], liposarcoma [13], leiomyosarcoma [14], salivary gland cancer [15], and chondroblastoma [16]. Cellular specific mechanism of TMEM16A is extensively reviewed and highlighted in various cancers [17,18]. Briefly, TMEM16A positively correlates with epidermal growth factor receptor (EGFR) expression in tumor development [2], and both TMEM16A and EGFR are found in NSCLC tissues. Tumor, node, metastasis (TNM) stage 3 + 4 primary NSCLC is positive for TMEM16A and EGFR [2]. Thus, TMEM16A is considered as a potential diagnostic marker for lung cancer. Treatment of TMEM16A inhibitor T16Ainh-A01 or knockdown of TMEM16A inhibits the cellular proliferation and invasion by attenuating EGFR phosphorylation in H1299 lung cancer cells [19]. Knocking down TMEM16A attenuates proliferation and migration by inhibiting phosphoinositide 3-kinase/protein kinase B (PI3K/PKB) and mitogen-activated protein kinase (MAPK) pathways in HCC, HepG2, and SMMC7721 cells [20]. Colorectal cancer (CRC) and HCT116 and DLD-1 cells also express abundant TMEM16A, which is a prognostic factor for patients with CRC [21]. MicroRNA-132 (miR-132) has been identified in nerve tissues of mice, humans, zebrafish, and cattle [22] and it functions as a tumor suppressor in various cancers to prevent metastasis and proliferation [23]. Attenuating TMEM16A through miR-132 decreases cellular proliferation, invasion, and liver metastasis [21]. In addition, bestrophin-1 is also considered as a putative calcium-activated chloride channel such as TMEM16A in epithelial cells such as the cystic fibrosis pancreatic duct cell line, CFPAC-1 [24,25,26,27,28] and it enhances calcium signaling and volume regulation in CRC T-84 cells by participating in proliferation [24]. Bestrophin-1 is also associated with the proliferation of oral squamous cell carcinoma (OSCC) HST-1 cells [27]. Although bestrophin-1 consists of calcium-activated chloride channels that are dependent on or independent of other proteins, it interacts with TMEM16A in normal tissues [29,30,31].

2.2. TMEM206

An acidic milieu is involved in various diseases, such as ischemia, cancer development, and inflammation [32]. Acid-sensitive chloride channels (also known as proton-activated chloride channels, PAC; TMEM206) are expressed in normal and malignant tissues. Protein profiling has revealed that colorectal, breast, and hepatic cancer cells have increased amounts of TMEM206, which plays a key role in cellular responses to acidic conditions [32,33]. Consistent with this concept, silencing TMEM206 attenuates acid-mediated cell death and alleviates acidosis-associated pathologies such as ischemic stroke. The cytoplasmic expression of TMEM206 is associated with CRC development and proliferation. The CRC cell lines SW480 and HCT-116 overexpress TMEM206, which results in enhanced cellular migration, invasion, and proliferation via AKT/ERK phosphorylation [32]. Although the precise mechanism of action of TMEM206 in other cancerous tissues remains unknown, TMEM206 could be considered as a diagnostic marker for CRC.

2.3. Calcium-Activated Chloride Channel Regulators

Calcium-activated chloride channel regulators (CLCAs) modulate chloride in epithelia, play critical roles in transporting electrolytes including chloride, modulate function of TMEM16A and its adhesion molecules, and negatively regulate cancer development. Nasopharyngeal, breast, and colorectal cancers have low levels of CLCAs [34,35,36,37,38]. The CLCA1 protein is primarily expressed in the small intestine, colon, and appendix. The expressions of CLCA1 and CLCA4 are decreased in intestinal tissues from patients with CRC, the CRC cell lines SW620 and LOVO, and in hormone receptor-positive breast cancer cell line MCF7 cells [35,37,38]. CLCA1 is negatively involved in the differentiation of intestinal Caco-2 cells [38]. Low levels of CLCA2 mRNA and protein have been identified in nasopharyngeal carcinoma (NPC) S18 and 5-8F cells, whereas overexpressed CLCA2 inhibits FAK/ERK signaling in these cells [34]. Transduction with p53 induces increased CLCA2 and inhibits the proliferation of breast cancer MCF10A and BT549 cells [36]. Overexpressed CLCA4 inhibits the epithelial-mesenchymal transition (EMT), which is involved in the migration and invasion of CRC cells, whereas CLCA4 depleted by shRNA enhances cellular migratory and invasive ability through enhanced EMT in human mammary epithelial cells [35,37]. Although further evidence is needed, CLCA levels could be considered as potential diagnostic biomarkers.

2.4. Chloride Intracellular Channels

Chloride intracellular channel 1 (CLIC1; also known as NCC27) belongs to the highly conserved CLIC family of chloride ion channels [39]. It can reside in the cytoplasm and temporarily in plasma and internal cell membranes [40]. CLIC1 participates in various cellular functions, including the maintenance of pH homeostasis, cell survival, cell cycle regulation, cell volume regulation, membrane potential modulation, and organelle acidification [40,41,42,43,44,45,46,47,48]. This channel is upregulated in various cancer type such as prostate [46], gallbladder (GBC) [48], colon cancer [47], gastric [49], clear cell renal cell carcinoma [50], and glioblastoma stem cells [51,52]. Overexpressed CLIC1 in patients with HCC [53] positively correlates with HCC proliferation and metastasis [54]. CLIC1 participates in hypoxia-induced colonic carcinoma metastasis via the MAPK/ERK pathway [45]. Moreover, CLIC1 is recruited to the plasma membrane in response to chemotaxis, such as directional treatment with epidermal growth factor (EGF) and mechanotaxis and its ectopic expression of CLIC1 enhances migratory apparatus such as lamellipodia and invadopodia [54]. Hypoxia-induced tumor cells possess irregular microvascular networks and blood flow [55] and can be transformed to promote cancer metastasis [56]. Mechanistically, limited blood perfusion or altered flow due to hypoxic conditions might contribute to the migration and invasion of cancer cells [57]. Upregulated CLIC1 expression correlates with lymph node metastasis and lymphatic invasion [49] as well as lung cancer migration and invasion [40]. Cell growth is promoted by CLIC1 via the MAPK/ERK pathway in prostate cancer [46] and CLIC1 is expressed in pancreatic ductal adenocarcinoma (PDAC) [58] where it plays an important role in promoting cancer cell survival, proliferation, and invasion [46,59]. In various regulatory processes involving CLIC1, small interfering (si)RNAs of CLIC1 induce the downregulation of cell proliferation, growth, and invasiveness of pancreatic cancer cell lines such as PANC-1 and MIAPaca-2 compared with control cells [58,60]. Furthermore, CLIC1 is associated with proteasome activator 28 β (PA28 β), and its specific siRNA downregulates CLIC1 in gastric cancer [61]. A regulatory volume decrease (RVD) is a critical process in cancer cell motility, such as migration and invasion [62]. The CLIC1 inhibitors IAA94 or CLIC1-specific siRNAs ameliorate the RVD and decrease the migration and invasion of CRC LOVO and HT-29 cell lines [45]. The expression of CLIC1 at the mRNA and protein levels is downregulated by miR-124 transfection in the hepatic cancer cell line HepG2 and by hsa-miR-372 transfection in the GBC cell lines G-415, OCUG-1, and SGC-996 [63,64]. The downregulation of CLIC1 reduces cell migration and invasion [63]. The siRNA of CLIC1 enhances expression of the tumor metastasis-related genes annexin A7 and gelsolin, the knockdown of which increases CLIC1 expression in mouse HCC Hca-F and Hca-P cell lines, suggesting that CLIC1 interacts with annexin A7 and gelsolin, and mediates tumor cell migration, invasion, and metastasis [65]. In addition, biguanide-related drugs such as metformin, morocydine, and proguanil inhibit CLIC1 current and dysregulate proliferation and invasiveness in glioblastoma stem cells [66]. Cells and tumors expressing Rab25 have abundant CLIC3 that co-localizes with active integrin α5β1 in ovarian cancer A2780 cells [67]. High CLIC4 expression in PDAC together with Indian hedgehog is a proposed metastatic marker of PDAC [68]. In contrast, CLIC2 is expressed in non-cancerous masses and is a potent regulator of tight junctions. The expression of CLIC2 and tight junction proteins is upregulated in non-cancer, compared with cancer cells, and CLIC2 regulates the formation of tight junction proteins such as claudin 1, claudin 5, zonula occludens-1, and occludin [69]. CLIC1 and other types of chloride channels could be potential treatment strategies for cancer and should be considered as novel diagnostic and therapeutic targets for prostate, gastric, gallbladder, colon, pancreas, lymphatic, and lung cancers.

2.5. Voltage-Gated Chloride Channels

Voltage-gated chloride channel 3 (CLC-3; also known as CLCN3) is expressed in cell membranes and intracellular vesicles where it exchanges chloride for hydrogen. CLC-3 protein is expressed in prostate carcinoma [70], nasopharyngeal [62], neuroendocrine [71], and brain cells [72] and is significantly overexpressed in HCC, compared with normal control tissues [73]. Moreover, upregulated CLC-3 is associated with HCC tumor size and prognosis [73]. Overexpressed CLC-3 protein participates in cell proliferation and migration. The regulation of cell volume by CLC-3 is involved in the development and metastasis of NPC and prostate cancer [62,70,74]. Signaling by Wnt/β-catenin contributes to metastasis and adhesion by regulating the EMT process in tumorigenesis [75,76]. The expression of CLC-3 is more abundant in tissues at the late stage of CRC and in the CRC LOVO and SW620 compared with that in normal cells. SiRNA-CLC-3 (siCLC-3) inhibits CRC cell viability, proliferation, and metastasis by inhibiting Wnt/β-catenin signaling, whereas the Wnt/β-catenin activator lithium chloride rescues the effect of siCLC-3 [77]. CLC-3 could be a prognostic marker for HCC, CRC, NPC, and prostate cancer. Patients with breast cancer are treated with tamoxifen, a non-steroidal anticancer agent [78] that inhibits the migration, chloride current, and volume regulatory mechanisms in HCC MHCC97H cells in vitro [79]. The activator of protein kinase C (PKC) phorbol-12-myristate-13 acetate (PMA) inhibits PKC expression in the presence of tamoxifen and reduces the migration of cells with CLC-3 knockdown, suggesting that CLC-3 is involved in the mechanism of anticancer drug and cellular volume regulation [80]. CLC-4 is expressed on the cell surface and intracellular endosomal membranes in the CRC cell lines RKO and LS174 [81]. The migration and invasion of CRC cells is reduced by CLC-4 siRNA or shRNA [82]. Incomplete glucose metabolism results in increased intracellular proton concentrations driven by CLC-4, which maintains a neutral intracellular pH and the essential proton extrusion mechanism [83]. The regulation of pH by CLC-4 in the endosomal compartment might participate in promoting invasive ability. Active acidification of large intracellular endosomal vesicles by a vacuolar H+-ATPase promotes proteolysis of the extracellular matrix, activates pro-cathepsin D which is activated when acidic condition is triggered, and facilitates proteolytic function on the basement membrane [82,84]. pH regulation in the cytosol and intracellular organelles in RKO cells overexpressing CLC-4 results in resistance to acid-induced cytotoxicity, which is similar to an acidic tumor microenvironment and the enhanced ability of colon cancer cells to migrate [82].

2.6. Cystic Fibrosis Transmembrane Conductance Regulator

Cystic fibrosis transmembrane conductance regulator (CFTR) is a cAMP-activated chloride channel that regulates the balance of electrolytes in the respiratory and endocrine systems, exocrine glands, and other tissues. Malfunctioning and/or abnormal expression of CFTR have been found in various types of cancer. The upregulated expression of CFTR is associated with an invasive phenotype in cervical and ovarian carcinomas [85,86]. Conversely, the mRNA and protein expression of CFTR are reduced in NPC 5-8F, 6-10B, and HNE-1, compared with normal cells, whereas CFTR knockdown increases NPC cell migration and invasion [87]. Enhanced CFTR protein expression in NPC 5-8F cells increases epithelial markers such as occludin and E-cadherin, and attenuates the mesenchymal marker smooth muscle actin [87]. In addition, protein expression of CFTR is decreased in CRC, compared with normal tissues [88]. However, CFTR mRNA overexpression decreases cell proliferation, migration, and invasion in the CRC cell lines HCT116 and CaCo-2 [88]. The results of studies on the roles of CFTR in cancer have been contradictory. Enhanced CFTR expression inhibits various cancerous processes such as EMT in breast carcinoma [89], lung cancer [90], NPC [87], endometrial carcinoma cells [91], prostate cancer [92], and intestinal carcinoma [93]. Cisplatin increases CFTR expression and enhances chemoresistance and the cell viability of prostate cancer tissues compared with chemo-sensitive prostate cancer tissues in vivo and LNCaP cells in vitro [94]. Nicotine is a potential cause of lung cancer and a progressive enhancer of adenocarcinoma cells that inhibits the CFTR protein expression in A549 cells [95]. Although the CFTR gene could act as a tumor suppressor, its roles in various cell types and cancer cells need to be defined.

2.7. Voltage-Dependent Anion Channels

The expression of voltage-dependent anion channels (VDACs) on the mitochondrial membrane of all eukaryotes, including mammals [96], is increased in various tumor tissues, such as carcinoma of the breast [97], colon [98], thyroid gland [99], lung [100], pancreas [101], and liver [102] compared with that in normal tissues. The VDAC1, 2, and 3 isotypes of these channels play different roles; VDAC1 and VDAC2 participate in pore formation within the mitochondrial membrane [96] and VDAC3 participates in the regulation of mitochondrial membrane potential [103]. The expression of VDAC is associated with neurodegenerative disorders and muscular and myocardial diseases including various types of cancers [104]. The progression of tumorigenesis is decreased in HeLa cells with depleted VDAC1 [105]. The expression of VDAC1 is more abundant in cancer, A549, and HeLa cells, than in normal WI-38 fibroblasts derived from lung tissue [106], HCC tissues, HepG2 and SMMC7721 cells, as well as lung adenocarcinoma tumors [107]. Small interfering RNA-VDAC1 and miR-7 downregulate cell growth, proliferation, migration, and invasion in HCC tissues [102], lung cancer A549 cells [100], and cervical cancer HeLa cells [105]. Furthermore, miR-490-3p is significantly associated with the carcinogenesis of various cancers [98], and it can regulate the growth and EMT of HCC cells [108] and the invasiveness of triple-negative breast cancer cells, MDA-MB-231, and MDA-MB-436 [109]. MiR-490-3p downregulates VDAC1 through the mammalian target of rapamycin (mTOR) pathway in CRC tissues and cell lines [98]. The expression of VDAC2 is upregulated in melanoma cells and HCC cell lines such as HepG2 [110] but downregulated in glioma stem cells [111] and it plays an anti-apoptotic role in primary cultured mouse embryonic fibroblasts. Although its different role of VDACs is defined, precise roles of VDAC family in different cancers remain unresolved and await identification in future studies.

2.8. Volume-Regulated Anion Channel

Volume regulation is critical function to maintain cellular fate. Volume-regulated anion channels (VRACs; also called volume-sensitive organic osmolyte anion channel or swelling-induced chloride current IClswell) are considered as regulatory channels of cellular volume [112]. VRAC is involved in the RVD and regulates proliferation of nasopharyngeal carcinoma cell [113,114,115], OSCC HST-1 cells [27], and gastric cancer [116]. Inhibited VRAC by 4-(2-Butyl-6,7-dichlor-2-cyclopentyl-indan-1-on-5-yl) oxybutyric acid (DCPIB) reduces proliferation, migration, and invasion of glioblastoma U251 and U87 cells [117]. Leucine-rich-repeat-containing 8A (LRRC8A, also called SWELL1) is component protein of VRAC. LRRC8A expresses in HCC tissues and induced cellular proliferation and migration in HCC SMMC-7721, Sk-hep-1, Huh7, and HCCLM3 cells [118]. Moreover, survival of cisplatin-resistant A549 cells or A2780 cells is modulated by the LRRC8A [119,120]. Although dual function of VRAC components LRRC8A and LRRC8D on drug resistance has been addressed [121], VRACs may be associated with chemo-resistant mechanism and are needed to verify its precise mechanism in various cancers.

2.9. Chloride-Bicarbonate Exchangers

Chloride-bicarbonate (CB) exchangers consist of solute carrier (SLC) families, including anion exchangers (AEs) and SLC26As. The CB exchangers mediate the electroneutral or electrogenic exchange of bicarbonate for chloride (respective stoichiometry of chloride: bicarbonate, 1:1 or 1:2) and are associated with the regulation of intracellular pH. The anion exchangers AE1, AE2, AE3, and AE4 [122,123,124,125] are expressed in various tissues and localize in the plasma membrane. Both AE1 and AE2 are involved in cancer whereas other AEs are unknown. Histologic findings have shown that AE1 is expressed in the cytoplasm of gastric cancer cells [126,127]. Knockdown of AE1 induces the release of p16INK4A and inhibits gastric cancer growth [126,128]. The expression of AE1 positively correlates with cancer size and metastasis [127]. Although the modulation of AE1 expression is poorly verified, miR-24-mediates AE1 attenuation in gastric cancer cells [129]. The expression of AE1 is associated with tumor progression through crosstalk with MAPK and hedgehog signaling pathways in esophageal carcinoma [130]. Although AE2 is expressed in most tissues, it has been addressed that the AE2 gene is highly expressed in HCC cells, gastric, and colorectal cancers [131,132,133,134,135]. An antisense oligonucleotide of AE2 inhibits HCC progression [135]. AE2 is also expressed in ovarian cancer and it participates in tumorigenesis through activation of the mTOR/p70S6K1 pathway [136]. AE2a that is localized in the Golgi apparatus is involved in the malignancy of SW-48 CRC cells [137]. We previously found that the anti-alcoholism agent disulfiram exerts anticancer effects and attenuates the membrane expression of AE2 and the supportive enzyme carbonic anhydrase XII through disturbed homeostatic pH regulation in lung cancer cells [138]. Our Western blotting findings in vitro also showed that disulfiram attenuated AE2 protein expression and CB exchanging activity in the breast cancer cell lines MDA-MB-231 and MCF-7 [138]. Although anti-cancer effect of disulfiram is addressed, the clinical relevance of AE2 in these cancers and the clinical effects of disulfiram should be verified more precisely in other types of cancer. The role of the SLC26 family in cancerous tissues or mechanisms is not well understood. Chondrodysplasias, chronic chloride diarrhea, and deafness in humans are linked to SLC26A2, SLC26A3, and SLC26A4, respectively [139]. SLC26A3 participates in chloride homeostasis and interactions with CFTR [140]. The expression of SLC26A3 is downregulated and modulated by the stomach-specific 18 kDa antrum mucosal protein in gastric cancer cells [141]. Analyses of gene sets and protein-protein networks have revealed a relationship between SLC26A6 and HCC [142]. However, further detailed investigation is needed to reveal whether any members of the SLC26A family have potential to serve as diagnostic or prognostic markers in cancerous tissues.

3. Perspectives

The modulation of chloride channels/transporters in cancers has been considered as challenging issues. Complicated ion movements, crosstalk among channels/transporters, difficulties with experimental implementation, and/or compensation of unidentified mechanisms hamper investigations into ion channels/transporters. For example, studies of intracellular ion channels, not plasma membrane-associated channels, have struggled with technical limitations in terms of verifying physiological roles [143].
Recently, advanced therapeutic candidates of chloride transport were reviewed in various organ diseases such as inflammatory lung disease, osteoporosis, dry eye disorders, hypertension, polycystic kidney disease, and kidney stone [112]. Although approved drugs or preclinical trials of chloride transporters are suggested [112], approaches in multiple cancers remain mostly unknown. Thus, this review would be beneficial to expand developed application of trials through an overview of chloride channel/transporter-associated cancers. Interestingly, several chloride channels/transporters are involved in at least two types of cancer (Figure 1 and Table 1). Although shared chloride channels might not reflect similarity among types of cancer, multiple facets should be considered to verify the roles of transporters and convergent regulation in normal and cancer tissues. Moreover, identifying crosstalk among cancers through shared chloride channels might provide valuable clues to metastatic cancer. Although accumulating information about chloride channels and transporters provides both challenges to investigator and potential therapeutic targets against cancer, mechanism of channels and transporters requires further verification. In addition, application of developed chloride transport modulators for cancers should be beneficial to verify off-target effect and toxicity on normal tissue for cancer treatment.

Author Contributions

H.J.K., P.C.-W.L. and J.H.H. contributed to the conceptualization and design of the review. H.J.K. collected the information, drafted the article, prepared the figure, and critically revised the manuscript for important intellectual content. J.H.H. and P.C.-W.L. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported a grant from The National Research Foundation of Korea (NRF) funded by the Korean government (MSIT; NRF-2019R1F1A1046785) to J.H.H. and (NRF- 2020R1A4A1016029) to P.C.-W.L.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Stauber, T.; Jentsch, T.J. Chloride in vesicular trafficking and function. Annu. Rev. Physiol. 2013, 75, 453–477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. He, Y.; Li, H.; Chen, Y.; Li, P.; Gao, L.; Zheng, Y.; Sun, Y.; Chen, J.; Qian, X. Expression of anoctamin 1 is associated with advanced tumor stage in patients with non-small cell lung cancer and predicts recurrence after surgery. Clin. Transl. Oncol. 2017, 19, 1091–1098. [Google Scholar] [CrossRef] [PubMed]
  3. Ardeleanu, C.; Arsene, D.; Hinescu, M.; Andrei, F.; Gutu, D.; Luca, L.; Popescu, L.M. Pancreatic expression of DOG1 a novel gastrointestinal stromal tumor (GIST) biomarker. Appl. Immunohistochem. Mol. Morphol. 2009, 17, 413–418. [Google Scholar] [CrossRef] [PubMed]
  4. Liu, W.; Lu, M.; Liu, B.G.; Huang, Y.; Wang, K.W. Inhibition of Ca2+-activated Cl channel ANO1/TMEM16A expression suppresses tumor growth and invasiveness in human prostate carcinoma. Cancer Lett. 2012, 326, 41–51. [Google Scholar] [CrossRef]
  5. Ubby, I.; Bussani, E.; Colonna, A.; Stacul, G.; Locatelli, M.; Scudieri, P.; Galietta, L.; Pagani, F. TMEM16A alternative splicing coordination in breast cancer. Mol. Cancer 2013, 12, 75. [Google Scholar] [CrossRef] [Green Version]
  6. Foda, A.M.; Mohamed, M.A. Aberrant expressions of c-KIT and DOG-1 in mucinous and nonmucinous colorectal carcinomas and relation to clinicopathologic features and prognosis. Ann. Diagn. Pathol. 2015, 19, 335–340. [Google Scholar] [CrossRef]
  7. Liu, F.; Cao, Q.H.; Lu, D.J.; Luo, B.; Lu, X.F.; Luo, R.C.; Wang, X.G. TMEM16A overexpression contributes to tumor invasion and poor prognosis of human gastric cancer through TGF-beta signaling. Oncotarget 2015, 6, 11585–11599. [Google Scholar] [CrossRef] [Green Version]
  8. Liu, J.; Liu, Y.; Ren, Y.G.; Kang, L.; Zhang, L.H. Transmembrane protein with unknown function 16A overexpression promotes glioma formation through the nuclear factor-.B signaling pathway. Mol. Med. Rep. 2014, 9, 1068–1074. [Google Scholar] [CrossRef] [Green Version]
  9. Lee, Y.S.; Lee, J.K.; Bae, Y.; Lee, B.S.; Kim, E.; Cho, C.H.; Ryoo, K.; Yoo, J.; Kim, C.H.; Yi, G.S.; et al. Suppression of 14-3-3 gamma-mediated surface expression of ANO1 inhibits cancer progression of glioblastoma cells. Sci. Rep. 2016, 6, 26413. [Google Scholar] [CrossRef] [Green Version]
  10. Shang, L.; Hao, J.J.; Zhao, X.K.; He, J.Z.; Shi, Z.Z.; Liu, H.J.; Wu, L.F.; Jiang, Y.Y.; Shi, F.; Yang, H.; et al. ANO1 protein as a potential biomarker for esophageal cancer prognosis and precancerous lesion development prediction. Oncotarget 2016, 7, 24374–24382. [Google Scholar] [CrossRef] [Green Version]
  11. Jia, L.H.; Liu, W.; Guan, L.Z.; Lu, M.; Wang, K.W. Inhibition of calcium-activated chloride channel ANO1/TMEM16A suppresses tumor growth and invasion in human lung cancer. PLoS ONE 2015, 10, e0136584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Deng, L.; Yang, J.H.; Chen, H.W.; Ma, B.; Pan, K.M.; Su, C.K.; Xu, F.F.; Zhang, J.H. Knockdown of TMEM16A suppressed MAPK and inhibited cell proliferation and migration in hepatocellular carcinoma. Oncotargets Ther. 2016, 9, 325–333. [Google Scholar] [CrossRef] [Green Version]
  13. Jung, I.; Gurzu, S.; Turdean, S.; Ciortea, D.; Sahlean, D.I.; Golea, M.; Bara, T. Relationship of endothelial area with VEGF-A, COX-2, maspin, c-KIT, and DOG-1 immunoreactivity in liposarcomas versus non-lipomatous soft tissue tumors. Int. J. Clin. Exp. Pathol. 2015, 8, 1776–1782. [Google Scholar] [PubMed]
  14. Sah, S.P.; McCluggage, W.G. DOG1 immunoreactivity in uterine leiomyosarcomas. J. Clin. Pathol. 2013, 66, 40–43. [Google Scholar] [CrossRef]
  15. Abd Raboh, N.M.; Hakim, S.A. Diagnostic role of DOG1 and p63 immunohistochemistry in salivary gland carcinomas. Int. J. Clin. Exp. Pathol. 2015, 8, 9214–9222. [Google Scholar]
  16. Akpalo, H.; Lange, C.; Zustin, J. Discovered on gastrointestinal stromal tumour 1 (DOG1): A useful immunohistochemical marker for diagnosing chondroblastoma. Histopathology 2012, 60, 1099–1106. [Google Scholar] [CrossRef]
  17. Wang, H.; Zou, L.; Ma, K.; Yu, J.; Wu, H.; Wei, M.; Xiao, Q. Cell-specific mechanisms of TMEM16A Ca2+-activated chloride channel in cancer. Mol. Cancer 2017, 16, 152. [Google Scholar] [CrossRef]
  18. Kunzelmann, K.; Ousingsawat, J.; Benedetto, R.; Cabrita, I.; Schreiber, R. Contribution of anoctamins to cell survival and cell death. Cancers 2019, 11, 382. [Google Scholar] [CrossRef] [Green Version]
  19. Hu, C.; Zhang, R.G.; Jiang, D.P. TMEM16A as a potential biomarker in the diagnosis and prognosis of lung cancer. Arch. Iran. Med. 2019, 22, 32–38. [Google Scholar]
  20. Zhang, C.T.; Liu, J.X.; Han, Z.Y.; Cui, X.; Peng, D.T.; Xing, Y.F. Inhibition of TMEM16A suppresses growth and induces apoptosis in hepatocellular carcinoma. Int. J. Clin. Oncol. 2020, 25, 1145–1154. [Google Scholar] [CrossRef]
  21. Mokutani, Y.; Uemura, M.; Munakata, K.; Okuzaki, D.; Haraguchi, N.; Takahashi, H.; Nishimura, J.; Hata, T.; Murata, K.; Takemasa, I.; et al. Down-regulation of microRNA-132 is associated with poor prognosis of colorectal cancer. Ann. Surg. Oncol. 2016, 23, S599–S608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Qian, Y.; Song, J.; Ouyang, Y.; Han, Q.; Chen, W.; Zhao, X.; Xie, Y.; Chen, Y.; Yuan, W.; Fan, C. Advances in Roles of miR-132 in the nervous system. Front. Pharmacol. 2017, 8, 770. [Google Scholar] [CrossRef] [PubMed]
  23. Wei, X.; Tan, C.; Tang, C.; Ren, G.; Xiang, T.; Qiu, Z.; Liu, R.; Wu, Z. Epigenetic repression of miR-132 expression by the hepatitis B virus x protein in hepatitis B virus-related hepatocellular carcinoma. Cell. Signal. 2013, 25, 1037–1043. [Google Scholar] [CrossRef] [PubMed]
  24. Spitzner, M.; Martins, J.R.; Soria, R.B.; Ousingsawat, J.; Scheidt, K.; Schreiber, R.; Kunzelmann, K. Eag1 and Bestrophin 1 are up-regulated in fast-growing colonic cancer cells. J. Biol. Chem. 2008, 283, 7421–7428. [Google Scholar] [CrossRef] [Green Version]
  25. Bharill, S.; Fu, Z.; Palty, R.; Isacoff, E.Y. Stoichiometry and specific assembly of Best ion channels. Proc. Natl. Acad. Sci. USA 2014, 111, 6491–6496. [Google Scholar] [CrossRef] [Green Version]
  26. Dam, V.S.; Boedtkjer, D.M.; Aalkjaer, C.; Matchkov, V. The bestrophin- and TMEM16A-associated Ca2+ -activated Cl channels in vascular smooth muscles. Channels 2014, 8, 361–369. [Google Scholar] [CrossRef] [Green Version]
  27. Yoshimoto, S.; Matsuda, M.; Kato, K.; Jimi, E.; Takeuchi, H.; Nakano, S.; Kajioka, S.; Matsuzaki, E.; Hirofuji, T.; Inoue, R.; et al. Volume-regulated chloride channel regulates cell proliferation and is involved in the possible interaction between TMEM16A and LRRC8A in human metastatic oral squamous cell carcinoma cells. Eur. J. Pharmacol. 2021, 895, 173881. [Google Scholar] [CrossRef]
  28. Marsey, L.L.; Winpenny, J.P. Bestrophin expression and function in the human pancreatic duct cell line, CFPAC-1. J. Physiol. 2009, 587, 2211–2224. [Google Scholar] [CrossRef]
  29. Kunzelmann, K.; Kongsuphol, P.; Aldehni, F.; Tian, Y.; Ousingsawat, J.; Warth, R.; Schreiber, R. Bestrophin and TMEM16-Ca2+ activated Cl channels with different functions. Cell Calcium 2009, 46, 233–241. [Google Scholar] [CrossRef]
  30. Barro-Soria, R.; Aldehni, F.; Almaca, J.; Witzgall, R.; Schreiber, R.; Kunzelmann, K. ER-localized bestrophin 1 activates Ca2+-dependent ion channels TMEM16A and SK4 possibly by acting as a counterion channel. Pflug. Arch. Eur. J. Phys. 2010, 459, 485–497. [Google Scholar] [CrossRef]
  31. Liu, Y.; Zhang, H.; Huang, D.; Qi, J.; Xu, J.; Gao, H.; Du, X.; Gamper, N.; Zhang, H. Characterization of the effects of Cl(-) channel modulators on TMEM16A and bestrophin-1 Ca2+ activated Cl channels. Pflug. Arch. 2015, 467, 1417–1430. [Google Scholar] [CrossRef] [PubMed]
  32. Zhao, J.B.; Zhu, D.H.; Zhang, X.P.; Zhang, Y.; Zhou, J.P.; Dong, M. TMEM206 promotes the malignancy of colorectal cancer cells by interacting with AKT and extracellular signal-regulated kinase signaling pathways. J. Cell. Physiol. 2019, 234, 10888–10898. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, L.; Liu, S.Y.; Yang, X.; Wang, Y.Q.; Cheng, Y.X. TMEM206 is a potential prognostic marker of hepatocellular carcinoma. Oncol. Lett. 2020, 20, 174. [Google Scholar] [CrossRef] [PubMed]
  34. Qiang, Y.Y.; Li, C.Z.; Sun, R.; Zheng, L.S.; Peng, L.X.; Yang, J.P.; Meng, D.F.; Lang, Y.H.; Mei, Y.; Xie, P.; et al. Along with its favorable prognostic role, CLCA2 inhibits growth and metastasis of nasopharyngeal carcinoma cells via inhibition of FAK/ERK signaling. J. Exp. Clin. Cancer Res. 2018, 37, 34. [Google Scholar] [CrossRef]
  35. Chen, H.; Liu, Y.; Jiang, C.D.; Chen, Y.M.; Li, H.; Liu, Q.A. Calcium-activated chloride channel A4 (CLCA4) plays inhibitory roles in invasion and migration through suppressing epithelial-mesenchymal transition via PI3K/AKT signaling in colorectal cancer. Med. Sci. Monit. 2019, 25, 4176–4185. [Google Scholar] [CrossRef]
  36. Walia, V.; Ding, M.; Kumar, S.; Nie, D.; Premkumar, L.S.; Elble, R.C. hCLCA2 is a p53-inducible inhibitor of breast cancer cell proliferation. Cancer Res. 2009, 69, 6624–6632. [Google Scholar] [CrossRef] [Green Version]
  37. Yu, Y.; Walia, V.; Elble, R.C. Loss of CLCA4 promotes epithelial-to-mesenchymal transition in breast cancer cells. PLoS ONE 2013, 8, e83943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Yang, B.; Cao, L.; Liu, B.; McCaig, C.D.; Pu, J. The transition from proliferation to differentiation in colorectal cancer is regulated by the calcium activated chloride channel A1. PLoS ONE 2013, 8, e60861. [Google Scholar] [CrossRef] [Green Version]
  39. Wang, W.; Wan, M.H.; Liao, D.J.; Peng, G.L.; Xu, X.; Yin, W.Q.; Guo, G.X.; Jiang, F.N.; Zhong, W.D.; He, J.X. Identification of potent chloride intracellular channel protein 1 inhibitors from traditional chinese medicine through structure-based virtual screening and molecular dynamics analysis. Biomed. Res. Int. 2017, 2017, 4751780. [Google Scholar] [CrossRef] [Green Version]
  40. Wang, W.; Xu, X.; Wang, W.J.; Shao, W.L.; Li, L.P.; Yin, W.Q.; Xiu, L.C.; Mo, M.C.; Zhao, J.; He, Q.Y.; et al. The expression and clinical significance of CLIC1 and HSP27 in lung adenocarcinoma. Tumor Biol. 2011, 32, 1199–1208. [Google Scholar] [CrossRef]
  41. Schlesinger, P.H.; Blair, H.C.; Teitelbaum, S.L.; Edwards, J.C. Characterization of the osteoclast ruffled border chloride channel and its role in bone resorption. J. Biol. Chem. 1997, 272, 18636–18643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Valenzuela, S.M.; Mazzanti, M.; Tonini, R.; Qiu, M.R.; Warton, K.; Musgrove, E.A.; Campbell, T.J.; Breit, S.N. The nuclear chloride ion channel NCC27 is involved in regulation of the cell cycle. J. Physiol. 2000, 529, 541–552. [Google Scholar] [CrossRef] [PubMed]
  43. Shi, Z.H.; Zhao, C.; Wu, H.; Wang, W.; Liu, X.M. CLIC1 protein A candidate prognostic biomarker for malignant-transformed hydatidiform Moles. Int. J. Gynecol. Cancer 2011, 21, 153–160. [Google Scholar] [CrossRef] [PubMed]
  44. Ma, P.F.; Chen, J.Q.; Wang, Z.; Liu, J.L.; Li, B.P. Function of chloride intracellular channel 1 in gastric cancer cells. World J. Gastroenterol. 2012, 18, 3070–3080. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, P.; Zhang, C.; Yu, P.W.; Tang, B.; Liu, T.; Cui, H.; Xu, J.H. Regulation of colon cancer cell migration and invasion by CLIC1-mediated RVD. Mol. Cell. Biochem. 2012, 365, 313–321. [Google Scholar] [CrossRef] [PubMed]
  46. Tian, Y.D.; Guan, Y.B.; Jia, Y.Y.; Meng, Q.J.; Yang, J.J. Chloride intracellular channel 1 regulates prostate cancer cell proliferation and migration through the MAPK/ERK pathway. Cancer Biother. Radiopharm. 2014, 29, 339–344. [Google Scholar] [CrossRef] [PubMed]
  47. Wang, P.; Zeng, Y.; Liu, T.; Zhang, C.; Yu, P.W.; Hao, Y.X.; Luo, H.X.; Liu, G. Chloride intracellular channel 1 regulates colon cancer cell migration and invasion through ROS/ERK pathway. World J. Gastroenterol. 2014, 20, 2071–2078. [Google Scholar] [CrossRef] [PubMed]
  48. Ding, Q.C.; Li, M.L.; Wu, X.S.; Zhang, L.; Wu, W.G.; Ding, Q.; Weng, H.; Wang, X.A.; Liu, Y.B. CLIC1 overexpression is associated with poor prognosis in gallbladder cancer. Tumor Biol. 2015, 36, 193–198. [Google Scholar] [CrossRef]
  49. Chen, C.D.; Wang, C.S.; Huang, Y.H.; Chien, K.Y.; Liang, Y.; Chen, W.J.; Lin, K.H. Overexpression of CLIC1 in human gastric carcinoma and its clinicopathological significance. Proteomics 2007, 7, 155–167. [Google Scholar] [CrossRef]
  50. Nesiu, A.; Cimpean, A.M.; Ceausu, R.A.; Adile, A.; Ioiart, I.; Porta, C.; Mazzanti, M.; Camerota, T.C.; Raica, M. Intracellular chloride ion channel protein-1 expression in clear cell renal cell carcinoma. Cancer Genomics Proteomics 2019, 16, 299–307. [Google Scholar] [CrossRef] [Green Version]
  51. Setti, M.; Savalli, N.; Osti, D.; Richichi, C.; Angelini, M.; Brescia, P.; Fornasari, L.; Carro, M.S.; Mazzanti, M.; Pelicci, G. Functional role of CLIC1 ion channel in glioblastoma-derived stem/progenitor cells. J. Natl. Cancer Inst. 2013, 105, 1644–1655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Peretti, M.; Raciti, F.M.; Carlini, V.; Verduci, I.; Sertic, S.; Barozzi, S.; Garre, M.; Pattarozzi, A.; Daga, A.; Barbieri, F.; et al. Mutual influence of ROS, pH, and CLIC1 membrane protein in the regulation of G1-S phase progression in human glioblastoma stem cells. Mol. Cancer Ther. 2018, 17, 2451–2461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Huang, J.S.; Chao, C.C.; Su, T.L.; Yeh, S.H.; Chen, D.S.; Chen, C.T.; Chen, P.J.; Jou, Y.S. Diverse cellular transformation capability of overexpressed genes in human hepatocellular carcinoma. Biochem. Biophys. Res. Commun. 2004, 315, 950–958. [Google Scholar] [CrossRef] [PubMed]
  54. Peng, J.M.; Lin, S.H.; Yu, M.C.; Hsieh, S.Y. CLIC1 recruits PIP5K1A/C to induce cell-matrix adhesions for tumor metastasis. J. Clin. Investig. 2021, 131. [Google Scholar] [CrossRef] [PubMed]
  55. Joyce, J.A.; Pollard, J.W. Microenvironmental regulation of metastasis. Nat. Rev. Cancer 2009, 9, 239–252. [Google Scholar] [CrossRef]
  56. Law, A.Y.; Wong, C.K. Stanniocalcin-2 promotes epithelial-mesenchymal transition and invasiveness in hypoxic human ovarian cancer cells. Exp. Cell Res. 2010, 316, 3425–3434. [Google Scholar] [CrossRef]
  57. Kokura, S.; Yoshida, N.; Imamoto, E.; Ueda, M.; Ishikawa, T.; Uchiyama, K.; Kuchide, M.; Naito, Y.; Okanoue, T.; Yoshikawa, T. Anoxia/reoxygenation down-regulates the expression of E-cadherin in human colon cancer cell lines. Cancer Lett. 2004, 211, 79–87. [Google Scholar] [CrossRef]
  58. Jia, N.N.; Dong, S.L.; Zhao, G.; Gao, H.; Li, X.Q.; Zhang, H.H. CLIC1 overexpression is associated with poor prognosis in pancreatic ductal adenocarcinomas. J. Cancer Res. Ther. 2016, 12, 892–896. [Google Scholar] [CrossRef]
  59. Zhang, S.; Wang, X.M.; Yin, Z.Y.; Zhao, W.X.; Zhou, J.Y.; Zhao, B.X.; Liu, P.G. Chloride intracellular channel 1 is overexpression in hepatic tumor and correlates with a poor prognosis. APMIS 2013, 121, 1047–1053. [Google Scholar] [CrossRef]
  60. Lu, J.H.; Dong, Q.; Zhang, B.T.; Wang, X.F.; Ye, B.; Zhang, F.; Song, X.L.; Gao, G.F.; Mu, J.S.; Wang, Z.; et al. Chloride intracellular channel 1 (CLIC1) is activated and functions as an oncogene in pancreatic cancer. Med. Oncol. 2015, 32, 171. [Google Scholar] [CrossRef]
  61. Zheng, D.L.; Huang, Q.L.; Zhou, F.; Huang, Q.J.; Lin, J.Y.; Lin, X. PA28 ss regulates cell invasion of gastric cancer via modulating the expression of chloride intracellular channel 1. J. Cell Biochem. 2012, 113, 1537–1546. [Google Scholar] [CrossRef] [PubMed]
  62. Mao, J.W.; Chen, L.X.; Xu, B.; Lijing, W.J.; Li, H.Z.; Guo, J.; Li, W.D.; Nie, S.H.; Jacob, T.J.C.; Wang, L.W. Suppression of ClC-3 channel expression reduces migration of nasopharyngeal carcinoma cells. Biochem. Pharmacol. 2008, 75, 1706–1716. [Google Scholar] [CrossRef] [PubMed]
  63. Yue, X.P.; Cui, Y.Y.; You, Q.; Lu, Y.X.; Zhang, J.F. MicroRNA-124 negatively regulates chloride intracellular channel 1 to suppress the migration and invasion of liver cancer cells. Oncol. Rep. 2019, 42, 1380–1390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Zhou, N.; Cheng, W.; Peng, C.; Liu, Y.; 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. 2017, 16, 7848–7854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Zhang, J.; Li, M.L.; Song, M.Y.; Chen, W.J.; Mao, J.; Song, L.; Wei, Y.Y.; Huang, Y.H.; Tang, J.W. Clic1 plays a role in mouse hepatocarcinoma via modulating Annexin A7 and Gelsolin in vitro and in vivo. Biomed. Pharmacother. 2015, 69, 416–419. [Google Scholar] [CrossRef]
  66. Barbieri, F.; Wurth, R.; Pattarozzi, A.; Verduci, I.; Mazzola, C.; Cattaneo, M.G.; Tonelli, M.; Solari, A.; Bajetto, A.; Daga, A.; et al. Inhibition of chloride intracellular channel 1 (CLIC1) as biguanide class-effect to impair human glioblastoma stem cell viability. Front. Pharmacol. 2018, 9, 899. [Google Scholar] [CrossRef] [Green Version]
  67. Dozynkiewicz, M.A.; Jamieson, N.B.; MacPherson, I.; Grindlay, J.; van den Berghe, P.V.E.; von Thun, A.; Morton, J.P.; 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 2012, 22, 131–145. [Google Scholar] [CrossRef] [Green Version]
  68. Zou, Q.; Yang, Z.L.; Li, D.Q.; Liu, Z.R.; Yuan, Y. Association of chloride intracellular channel 4 and Indian hedgehog proteins with survival of patients with pancreatic ductal adenocarcinoma. Int. J. Exp. Pathol. 2016, 97, 422–429. [Google Scholar] [CrossRef]
  69. Ueno, Y.; Ozaki, S.; Umakoshi, A.; Yano, H.; Choudhury, M.E.; 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 2019, 7, e1593775. [Google Scholar] [CrossRef] [Green Version]
  70. Lemonnier, L.; Shuba, Y.; Crepin, A.; Roudbaraki, M.; Slomianny, C.; Mauroy, B.; Nilius, B.; Prevarskaya, N.; Skryma, R. Bcl-2-dependent modulation of swelling-activated Cl- current and ClC-3 expression in human prostate cancer epithelial cells. Cancer Res. 2004, 64, 4841–4848. [Google Scholar] [CrossRef] [Green Version]
  71. Weylandt, K.H.; Nebrig, M.; Jansen-Rosseck, N.; Amey, J.S.; Carmena, D.; Wiedenmann, B.; Higgins, C.F.; Sardini, A. CIC-3 expression enhances etoposide resistance by increasing acidification of the late endocytic compartment. Mol. Cancer Ther. 2008, 7, 2261. [Google Scholar] [CrossRef] [Green Version]
  72. Sontheimer, H. An unexpected role for ion channels in brain tumor metastasis. Exp. Biol. Med. 2008, 233, 779–791. [Google Scholar] [CrossRef] [Green Version]
  73. Cheng, W.; Zheng, S.T.; Li, L.; Zhou, Q.; Zhu, H.P.; Hu, J.; Luo, H.B. Chloride channel 3 (CIC-3) predicts the tumor size in hepatocarcinoma. Acta Histochem. 2019, 121, 284–288. [Google Scholar] [CrossRef] [PubMed]
  74. Xu, B.; Mao, J.W.; Wang, L.W.; Zhu, L.Y.; Li, H.Z.; Wang, W.Z.; Jin, X.B.; Zhu, J.Y.; Chen, L.X. ClC-3 chloride channels are essential for cell proliferation and cell cycle progression in nasopharyngeal carcinoma cells. Acta Biochim. Biophys. Sin. 2010, 42, 370–380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Fodde, R.; Brabletz, T. Wnt/beta-catenin signaling in cancer stemness and malignant behavior. Curr. Opin. Cell Biol. 2007, 19, 150–158. [Google Scholar] [CrossRef] [PubMed]
  76. Cai, J.C.; Guan, H.Y.; Fang, L.S.; Yang, Y.; Zhu, X.; Yuan, J.; Wu, J.H.; Li, M.F. MicroRNA-374a activates Wnt/beta-catenin signaling to promote breast cancer metastasis. J. Clin. Investig. 2013, 123, 566–579. [Google Scholar] [CrossRef] [Green Version]
  77. Mu, H.L.; Mu, L.J.; Gao, J.F. Suppression of CLC-3 reduces the proliferation, invasion and migration of colorectal cancer through Wnt/beta-catenin signaling pathway. Biochem. Biophys. Res. Commun. 2020, 533, 1240–1246. [Google Scholar] [CrossRef]
  78. Hughes-Davies, L.; Caldas, C.; Wishart, G.C. Tamoxifen: The drug that came in from the cold. Br. J. Cancer 2009, 101, 875–878. [Google Scholar] [CrossRef] [Green Version]
  79. Mao, J.W.; Xu, B.; Li, H.Z.; Chen, L.X.; Jin, X.B.; Zhu, J.Y.; Wang, W.Z.; Zhu, L.Y.; Zuo, W.H.; Chen, W.Q.; et al. Lack of association between stretch-activated and volume-activated Cl currents in hepatocellular carcinoma cells. J. Cell. Physiol. 2011, 226, 1176–1185. [Google Scholar] [CrossRef]
  80. Mao, J.W.; Yuan, J.; Wang, L.W.; Zhang, H.F.; Jin, X.B.; Zhu, J.Y.; Li, H.Z.; Xu, B.; Chen, L.X. Tamoxifen inhibits migration of estrogen receptor-negative hepatocellular carcinoma cells by blocking the swelling-activated chloride current. J. Cell. Physiol. 2013, 228, 991–1001. [Google Scholar] [CrossRef]
  81. Mohammad-Panah, R.; Ackerley, C.; Rommens, J.; Choudhury, M.; Wang, Y.C.; Bear, C.E. The chloride channel ClC-4 co-localizes with cystic fibrosis transmembrane conductance regulator and may mediate chloride flux across the apical membrane of intestinal epithelia. J. Biol. Chem. 2002, 277, 566–574. [Google Scholar] [CrossRef] [Green Version]
  82. Ishiguro, T.; Avila, H.; Lin, S.Y.; Nakamura, T.; Yamamoto, M.; Boyd, D.D. Gene trapping identifies chloride channel 4 as a novel inducer of colon cancer cell migration, invasion and metastases. Br. J. Cancer 2010, 102, 774–782. [Google Scholar] [CrossRef] [Green Version]
  83. Gatenby, R.A.; Gillies, R.J. Why do cancers have high aerobic glycolysis? Nat. Rev. Cancer 2004, 4, 891–899. [Google Scholar] [CrossRef] [PubMed]
  84. Montcourrier, P.; Mangeat, P.H.; Valembois, C.; Salazar, G.; Sahuquet, A.; Duperray, C.; Rochefort, H. Characterization of very acidic phagosomes in breast-cancer cells and their association with invasion. J. Cell Sci. 1994, 107, 2381–2391. [Google Scholar] [CrossRef] [PubMed]
  85. Xu, J.; Lin, L.B.; Yong, M.; Dong, X.J.; Yu, T.H.; Hu, L.N. Adenovirus-mediated overexpression of cystic fibrosis transmembrane conductance regulator enhances invasiveness and motility of serous ovarian cancer cells. Mol. Med. Rep. 2016, 13, 265–272. [Google Scholar] [CrossRef] [PubMed]
  86. Peng, X.; Wu, Z.; Yu, L.; Li, J.K.; Xu, W.M.; Chan, H.C.; Zhang, Y.; Hu, L.N. Overexpression of cystic fibrosis transmembrane conductance regulator (CFTR) is associated with human cervical cancer malignancy, progression and prognosis. Gynecol. Oncol. 2012, 125, 470–476. [Google Scholar] [CrossRef] [PubMed]
  87. Tu, Z.W.; Chen, Q.; Zhang, J.T.; Jiang, X.H.; Xia, Y.F.; Chan, H.C. CFTR is a potential marker for nasopharyngeal carcinoma prognosis and metastasis. Oncotarget 2016, 7, 76955–76965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Liu, C.; Song, C.; Li, J.X.; Sun, Q. CFTR functions as a tumor suppressor and is regulated by DNA methylation in colorectal cancer. Cancer Manag. Res. 2020, 12, 4261–4270. [Google Scholar] [CrossRef]
  89. Zhang, J.T.; Jiang, X.H.; Xie, C.; Cheng, H.; Dong, J.D.; Wang, Y.; Fok, K.L.; Zhang, X.H.; Sun, T.T.; Tsang, L.L.; et al. Downregulation of CFTR promotes epithelial-to-mesenchymal transition and is associated with poor prognosis of breast cancer. BBA Mol. Cell Res. 2013, 1833, 2961–2969. [Google Scholar] [CrossRef] [Green Version]
  90. Li, J.; Zhang, J.T.; Jiang, X.H.; Shi, X.S.; Shen, J.F.; Feng, F.L.; Chen, J.Y.; Liu, G.H.; He, P.; Jiang, J.H.; et al. The cystic fibrosis transmembrane conductance regulator as a biomarker in non-small cell lung cancer. Int. J. Oncol. 2015, 46, 2107–2115. [Google Scholar] [CrossRef] [Green Version]
  91. Xia, X.; Wang, J.; Liu, Y.; Yue, M. Lower cystic fibrosis transmembrane conductance regulator (CFTR) promotes the proliferation and migration of endometrial carcinoma. Med. Sci. Monit. 2017, 23, 966–974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Xie, C.; Jiang, X.H.; Zhang, J.T.; Sun, T.T.; Dong, J.D.; Sanders, A.J.; Diao, R.Y.; Wang, Y.; Fok, K.L.; Tsang, L.L.; et al. CFTR suppresses tumor progression through miR-193b targeting urokinase plasminogen activator (uPA) in prostate cancer. Oncogene 2013, 32, 2282–2291. [Google Scholar] [CrossRef] [PubMed]
  93. Than, B.L.N.; Linnekamp, J.F.; Starr, T.K.; Largaespada, D.A.; 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 2016, 35, 4191–4199. [Google Scholar] [CrossRef] [PubMed]
  94. Zhu, Q.; Li, H.; Liu, Y.; Jiang, L. Knockdown of CFTR enhances sensitivity of prostate cancer cells to cisplatin via inhibition of autophagy. Neoplasma 2017, 64, 709–717. [Google Scholar] [CrossRef] [Green Version]
  95. Li, H.; Ma, N.X.; Wang, J.; Wang, Y.; Yuan, C.; Wu, J.; Luo, M.H.; Yang, J.L.; Chen, J.; Shi, J.; et al. Nicotine induces progressive properties of lung adenocarcinoma A549 cells by inhibiting cystic fibrosis transmembrane conductance regulator (CFTR) expression and plasma membrane localization. Technol. Cancer Res. Treat. 2018, 17, 1533033818809984. [Google Scholar] [CrossRef]
  96. Jozwiak, P.; Ciesielski, P.; Forma, E.; Kozal, K.; Wojcik-Krowiranda, K.; Cwonda, L.; Bienkiewicz, A.; Brys, M.; Krzeslak, A. Expression of voltage-dependent anion channels in endometrial cancer and its potential prognostic significance. Tumour Biol. 2020, 42, 1010428320951057. [Google Scholar] [CrossRef]
  97. Yang, G.; Zhou, D.; Li, J.; Wang, W.; Zhong, W.; Fan, W.; Yu, M.; Cheng, H. VDAC1 is regulated by BRD4 and contributes to JQ1 resistance in breast cancer. Oncol. Lett. 2019, 18, 2340–2347. [Google Scholar] [CrossRef]
  98. Liu, X.; He, B.; Xu, T.; Pan, Y.; Hu, X.; Chen, X.; Wang, S. MiR-490-3p functions as a tumor suppressor by inhibiting oncogene VDAC1 expression in colorectal cancer. J. Cancer 2018, 9, 1218–1230. [Google Scholar] [CrossRef] [Green Version]
  99. Ko, J.H.; Gu, W.; Lim, I.; Zhou, T.; Bang, H. Expression profiling of mitochondrial voltage-dependent anion channel-1 associated genes predicts recurrence-free survival in human carcinomas. PLoS ONE 2014, 9, e110094. [Google Scholar] [CrossRef] [Green Version]
  100. Arif, T.; Vasilkovsky, L.; Refaely, Y.; Konson, A.; Shoshan-Barmatz, V. Silencing VDAC1 expression by siRNA inhibits cancer cell proliferation and tumor growth in vivo. Mol. Ther. Nucleic Acids 2014, 3, e159. [Google Scholar] [CrossRef]
  101. Wang, W.; Zhang, T.; Zhao, W.; Xu, L.; Yang, Y.; Liao, Q.; Zhao, Y. A single talent immunogenic membrane antigen and novel prognostic predictor: Voltage-dependent anion channel 1 (VDAC1) in pancreatic cancer. Sci. Rep. 2016, 6, 33648. [Google Scholar] [CrossRef] [Green Version]
  102. Wang, F.R.; Qiang, Y.; Zhu, L.R.; Jiang, Y.S.; Wang, Y.D.; Shao, X.; Yin, L.; Chen, J.H.; Chen, Z. MicroRNA-7 downregulates the oncogene VDAC1 to influence hepatocellular carcinoma proliferation and metastasis. Tumor Biol. 2016, 37, 10235–10246. [Google Scholar] [CrossRef]
  103. Maldonado, E.N.; Lemasters, J.J. Warburg revisited: Regulation of mitochondrial metabolism by voltage-dependent anion channels in cancer cells. J. Pharmacol. Exp. Ther. 2012, 342, 637–641. [Google Scholar] [CrossRef] [Green Version]
  104. Shoshan-Barmatz, V.; De Pinto, V.; Zweckstetter, M.; Raviv, Z.; Keinan, N.; Arbel, N. VDAC, a multi-functional mitochondrial protein regulating cell life and death. Mol. Asp. Med. 2010, 31, 227–285. [Google Scholar] [CrossRef] [PubMed]
  105. Koren, I.; Raviv, Z.; Shoshan-Barmatz, V. Downregulation of voltage-dependent anion channel-1 expression by RNA interference prevents cancer cell growth in vivo. Cancer Biol. Ther. 2010, 9, 1046–1052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Simamura, E.; Hirai, K.; Shimada, H.; Koyama, J.; Niwa, Y.; Shimizu, S. Furanonaphthoquinones cause apoptosis of cancer cells by inducing the production of reactive oxygen species by the mitochondrial voltage-dependent anion channel. Cancer Biol. Ther. 2006, 5, 1523–1529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Pernemalm, M.; De Petris, L.; Branca, R.M.; Forshed, J.; Kanter, L.; Soria, J.C.; Girard, P.; Validire, P.; Pawitan, Y.; van den Oord, J.; et al. Quantitative proteomics profiling of primary lung adenocarcinoma tumors reveals functional perturbations in tumor metabolism. J. Proteome Res. 2013, 12, 3934–3943. [Google Scholar] [CrossRef] [PubMed]
  108. Zhang, L.Y.; Liu, M.; Li, X.; Tang, H. miR-490-3p modulates cell growth and epithelial to mesenchymal transition of hepatocellular carcinoma cells by targeting endoplasmic reticulum-Golgi intermediate compartment protein 3 (ERGIC3). J. Biol. Chem. 2013, 288, 4035–4047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Jia, Z.; Liu, Y.; Gao, Q.; Han, Y.; Zhang, G.; Xu, S.; Cheng, K.; Zou, W. miR-490-3p inhibits the growth and invasiveness in triple-negative breast cancer by repressing the expression of TNKS2. Gene 2016, 593, 41–47. [Google Scholar] [CrossRef]
  110. Maldonado, E.N.; Sheldon, K.L.; DeHart, D.N.; Patnaik, J.; Manevich, Y.; Townsend, D.M.; Bezrukov, S.M.; Rostovtseva, T.K.; Lemasters, J.J. Voltage-dependent anion channels modulate mitochondrial metabolism in cancer cells: Regulation by free tubulin and erastin. J. Biol. Chem. 2013, 288, 11920–11929. [Google Scholar] [CrossRef] [Green Version]
  111. Zhou, K.; Yao, Y.L.; He, Z.C.; Chen, C.; Zhang, X.N.; Yang, K.D.; Liu, Y.Q.; Liu, Q.; Fu, W.J.; Chen, Y.P.; et al. VDAC2 interacts with PFKP to regulate glucose metabolism and phenotypic reprogramming of glioma stem cells. Cell Death Dis. 2018, 9, 988. [Google Scholar] [CrossRef]
  112. Verkman, A.S.; Galietta, L.J.V. Chloride transport modulators as drug candidates. Am. J. Physiol. Cell Physiol. 2021, 321, C932–C946. [Google Scholar] [CrossRef] [PubMed]
  113. Chen, L.; Wang, L.; Zhu, L.; Nie, S.; Zhang, J.; Zhong, P.; Cai, B.; Luo, H.; Jacob, T.J. Cell cycle-dependent expression of volume-activated chloride currents in nasopharyngeal carcinoma cells. Am. J. Physiol. Cell Physiol. 2002, 283, C1313–C1323. [Google Scholar] [CrossRef] [PubMed]
  114. Wang, L.W.; Chen, L.X.; Zhu, L.Y.; Rawle, M.; Nie, S.H.; Zhang, J.; Ping, Z.; Cai, K.R.; Jacob, T.J.C. Regulatory volume decrease is actively modulated during the cell cycle. J. Cell. Physiol. 2002, 193, 110–119. [Google Scholar] [CrossRef] [PubMed]
  115. Chen, L.X.; Zhu, L.Y.; Jacob, T.J.; Wang, L.W. Roles of volume-activated Cl- currents and regulatory volume decrease in the cell cycle and proliferation in nasopharyngeal carcinoma cells. Cell Prolif. 2007, 40, 253–267. [Google Scholar] [CrossRef]
  116. 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 2021, 24, 1063–1075. [Google Scholar] [CrossRef]
  117. Wong, R.; Chen, W.; Zhong, X.; Rutka, J.T.; Feng, Z.P.; Sun, H.S. Swelling-induced chloride current in glioblastoma proliferation, migration, and invasion. J. Cell. Physiol. 2018, 233, 363–370. [Google Scholar] [CrossRef]
  118. Lu, P.; Ding, Q.; Li, X.; Ji, X.; Li, L.; Fan, Y.; Xia, Y.; Tian, D.; Liu, M. SWELL1 promotes cell growth and metastasis of hepatocellular carcinoma in vitro and in vivo. EBioMedicine 2019, 48, 100–116. [Google Scholar] [CrossRef] [Green Version]
  119. Min, X.J.; Li, H.; Hou, S.C.; He, W.; Liu, J.; Hu, B.; Wang, J. Dysfunction of volume-sensitive chloride channels contributes to cisplatin resistance in human lung adenocarcinoma cells. Exp. Biol. Med. 2011, 236, 483–491. [Google Scholar] [CrossRef]
  120. Sorensen, B.H.; Dam, C.S.; Sturup, S.; Lambert, I.H. Dual role of LRRC8A-containing transporters on cisplatin resistance in human ovarian cancer cells. J. Inorg. Biochem. 2016, 160, 287–295. [Google Scholar] [CrossRef]
  121. Planells-Cases, R.; Lutter, D.; Guyader, C.; Gerhards, N.M.; Ullrich, F.; Elger, D.A.; Kucukosmanoglu, A.; Xu, G.; Voss, F.K.; Reincke, S.M.; et al. Subunit composition of VRAC channels determines substrate specificity and cellular resistance to Pt-based anti-cancer drugs. EMBO J. 2015, 34, 2993–3008. [Google Scholar] [CrossRef] [PubMed]
  122. Mueller, T.J.; Morrison, M. Detection of a variant of protein 3, the major transmembrane protein of the human erythrocyte. J. Biol. Chem. 1977, 252, 6573–6576. [Google Scholar] [CrossRef]
  123. Demuth, D.R.; Showe, L.C.; Ballantine, M.; Palumbo, A.; Fraser, P.J.; Cioe, L.; Rovera, G.; Curtis, P.J. Cloning and structural characterization of a human non-erythroid band 3-like protein. EMBO J. 1986, 5, 1205–1214. [Google Scholar] [CrossRef] [PubMed]
  124. Kopito, R.R.; Lee, B.S.; Simmons, D.M.; Lindsey, A.E.; Morgans, C.W.; Schneider, K. Regulation of intracellular pH by a neuronal homolog of the erythrocyte anion exchanger. Cell 1989, 59, 927–937. [Google Scholar] [CrossRef]
  125. Tsuganezawa, H.; Kobayashi, K.; Iyori, M.; Araki, T.; Koizumi, A.; Watanabe, S.I.; Kaneko, A.; Fukao, T.; Monkawa, T.; Yoshida, T.; et al. A new member of the HCO 3-transporter superfamily is an apical anion exchanger of beta-intercalated cells in the kidney. J. Biol. Chem. 2001, 276, 8180–8189. [Google Scholar] [CrossRef] [Green Version]
  126. Shen, W.W.; Wu, J.; Cai, L.; Liu, B.Y.; Gao, Y.; Chen, G.Q.; Fu, G.H. Expression of anion exchanger 1 sequestrates p16 in the cytoplasm in gastric and colonic adenocarcinoma. Neoplasia 2007, 9, 812–819. [Google Scholar] [CrossRef] [Green Version]
  127. Xu, W.Q.; Song, L.J.; Liu, Q.; Zhao, L.; Zheng, L.; Yan, Z.W.; Fu, G.H. Expression of anion exchanger 1 is associated with tumor progress in human gastric cancer. J. Cancer Res. Clin. 2009, 135, 1323–1330. [Google Scholar] [CrossRef] [PubMed]
  128. Suo, W.H.; Zhang, N.; Wu, P.P.; Zhao, L.; Song, L.J.; Shen, W.W.; Zheng, L.; Tao, J.; Long, X.D.; Fu, G.H. Anti-tumour effects of small interfering RNA targeting anion exchanger 1 in experimental gastric cancer. Br. J. Pharmacol. 2012, 165, 135–147. [Google Scholar] [CrossRef] [Green Version]
  129. Wu, J.; Zhang, Y.C.; Suo, W.H.; Liu, X.B.; Shen, W.W.; Tian, H.; Fu, G.H. Induction of anion exchanger-1 translation and its opposite roles in the carcinogenesis of gastric cancer cells and differentiation of K562 cells. Oncogene 2010, 29, 1987–1996. [Google Scholar] [CrossRef] [Green Version]
  130. Shiozaki, A.; Kudou, M.; Ichikawa, D.; Shimizu, H.; Arita, T.; Kosuga, T.; Konishi, H.; Komatsu, S.; Fujiwara, H.; Okamoto, K.; et al. Expression and role of anion exchanger 1 in esophageal squamous cell carcinoma. Oncotarget 2017, 8, 17921–17935. [Google Scholar] [CrossRef] [Green Version]
  131. Wu, T.T.; Hsieh, Y.H.; Wu, C.C.; Tsai, J.H.; Hsieh, Y.S.; Huang, C.Y.; Liu, J.Y. Overexpression of anion exchanger 2 in human hepatocellular carcinoma. Chin. J. Physiol. 2006, 49, 192–198. [Google Scholar] [PubMed]
  132. Song, L.J.; Liu, R.J.; Zeng, Z.; Alper, S.L.; Cui, H.J.; Lu, Y.; Zheng, L.; Yan, Z.W.; Fu, G.H. Gastrin inhibits a novel, pathological colon cancer signaling pathway involving EGR1, AE2, and P-ERK. J. Mol. Med. 2012, 90, 707–718. [Google Scholar] [CrossRef] [PubMed]
  133. Wang, T.; Zhao, L.; Yang, Y.; Tian, H.; Suo, W.H.; Yan, M.; Fu, G.H. EGR1 is critical for gastrin-dependent upregulation of anion exchanger 2 in gastric cancer cells. FEBS J. 2013, 280, 174–183. [Google Scholar] [CrossRef]
  134. Yang, Y.; Wu, P.P.; Wu, J.; Shen, W.W.; Wu, Y.L.; Fu, A.F.; Zheng, L.; Jin, X.L.; Fu, G.H. Expression of anion exchanger 2 in human gastric cancer. Exp. Oncol. 2008, 30, 81–87. [Google Scholar]
  135. Hwang, J.M.; Kao, S.H.; Hsieh, Y.H.; Li, K.L.; Wang, P.H.; Hsu, L.S.; Liu, J.Y. Reduction of anion exchanger 2 expression induces apoptosis of human hepatocellular carcinoma cells. Mol. Cell. Biochem. 2009, 327, 135–144. [Google Scholar] [CrossRef] [PubMed]
  136. Zhang, L.J.; Lu, R.; Song, Y.N.; Zhu, J.Y.; Xia, W.; Zhang, M.; Shao, Z.Y.; Huang, Y.; Zhou, Y.; Zhang, H.; et al. Knockdown of anion exchanger 2 suppressed the growth of ovarian cancer cells via mTOR/p70S6K1 signaling. Sci. Rep. 2017, 7, 6362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Khosrowabadi, E.; Rivinoja, A.; Risteli, M.; Tuomisto, A.; Salo, T.; Makinen, M.J.; Kellokumpu, S. SLC4A2 anion exchanger promotes tumour cell malignancy via enhancing net acid efflux across golgi membranes. Cell. Mol. Life Sci. 2021, 78, 6283–6304. [Google Scholar] [CrossRef] [PubMed]
  138. Hwang, S.; Shin, D.M.; Hong, J.H. Drug repurposing as an antitumor agent: Disulfiram-mediated carbonic anhydrase 12 and anion exchanger 2 modulation to inhibit cancer cell migration. Molecules 2019, 24, 3409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Cordat, E.; Reithmeier, R.A. Structure, function, and trafficking of SLC4 and SLC26 anion transporters. Curr. Top. Membr. 2014, 73, 1–67. [Google Scholar] [CrossRef]
  140. Ko, S.B.; Shcheynikov, N.; Choi, J.Y.; Luo, X.; Ishibashi, K.; Thomas, P.J.; Kim, J.Y.; Kim, K.H.; Lee, M.G.; Naruse, S.; et al. A molecular mechanism for aberrant CFTR-dependent HCO3− transport in cystic fibrosis. EMBO J. 2002, 21, 5662–5672. [Google Scholar] [CrossRef] [PubMed]
  141. Di Stadio, C.S.; Altieri, F.; Miselli, G.; Elce, A.; Severino, V.; Chambery, A.; Quagliariello, V.; Villano, V.; de Dominicis, G.; Rippa, E.; et al. AMP18 interacts with the anion exchanger SLC26A3 and enhances its expression in gastric cancer cells. Biochimie 2016, 121, 151–160. [Google Scholar] [CrossRef] [PubMed]
  142. Zhu, Y.; Huang, Y.; Chen, L.; Guo, L.; Wang, L.; Li, M.; Liang, Y. Up-regulation of SLC26A6 in hepatocellular carcinoma and its diagnostic and prognostic significance. Crit. Rev. Eukaryot. Gene Expr. 2021, 31, 79–94. [Google Scholar] [CrossRef] [PubMed]
  143. Gururaja Rao, S.; Patel, N.J.; Singh, H. Intracellular chloride channels: Novel biomarkers in diseases. Front. Physiol. 2020, 11, 96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Schema of chloride channel-associated cancers. Chloride channels are involved in at least two types of cancers. Identification of shared chloride channels might provide clues to identify metastatic cancer. VDAC1: Voltage-dependent anion channel 1, TMEM: Transmembrane member, CLC: Voltage-gated chloride channels, CLIC: Chloride intracellular channels, CFTR: Cystic fibrosis transmembrane conductance regulator.
Figure 1. Schema of chloride channel-associated cancers. Chloride channels are involved in at least two types of cancers. Identification of shared chloride channels might provide clues to identify metastatic cancer. VDAC1: Voltage-dependent anion channel 1, TMEM: Transmembrane member, CLC: Voltage-gated chloride channels, CLIC: Chloride intracellular channels, CFTR: Cystic fibrosis transmembrane conductance regulator.
Cancers 14 00856 g001
Table 1. Diseases-related chloride channels/transporters and their functions.
Table 1. Diseases-related chloride channels/transporters and their functions.
DiseasesChloride Channels/TransportersFunctionReference
Lung cancerTMEM16ACell proliferation and development to malignant tumor
Positive correlation between TMEM16A and EGFR
[2,11,19]
TMEM206Enhanced cellular migration, invasion, and proliferation[32]
CLIC1Enhanced cellular migration and invasion[40]
CFTRInhibited various cancer-related processes, such as EMT[90,95]
VDAC1Cell growth, proliferation, migration, and invasion[100,107]
Pancreatic cancerTMEM16ACell proliferation and development to malignant tumor[3]
CLIC1Cell proliferation, growth, and invasiveness[58,60]
CLIC4Proposed metastatic marker[68]
VDAC1Cell growth, invasion, and migration[101]
Hepatic cancerTMEM16ACell proliferation and development to malignant tumor[12,20]
CLIC1Promoted cancer cell survival, proliferation, and invasion[53,54,59,63,65]
CLIC2Correlated with the tight junction[69]
CLC3Related to tumor size and prognosis marker
Cell volume regulation
[73,79,80]
VDAC1Regulated cell growth and EMT
Cell growth, proliferation, migration, and invasion
[102,108]
VDAC3Regulation of mitochondrial membrane potential[103]
AE2Inhibited proliferation and viability of cancer cells[131,135]
SLC26A6Diagnostic or prognostic biomarker for cancer[142]
Nasopharyngeal cancerCLCA2Negatively regulate cancer development
Regulate FAK/ERK signaling
[34]
CLC3Correlation between cell volume regulation and cancer development and metastasis[62,74]
CFTREnhanced cell migration and invasion
Regulate EMT
[87]
VRACRegulates proliferation[115]
Breast cancerTMEM16ACell proliferation and development to malignant tumor[5]
CLCA2Negatively regulate cancer development[36]
CLCA4Negatively regulate cancer development
Decreased EMT
[37]
CFTRInhibited various cancer-related processes, such as EMT[89]
VDAC1Promoted cell proliferation[97]
Colorectal cancerTMEM16ACell proliferation and development to malignant tumor[6,21]
Bestrophin-1Involved in cellular proliferation[24]
CLCA1Negatively regulate cancer development[38]
CLCA4Negatively regulate cancer development
Decreased EMT
[35]
CLIC1Enhanced cellular migration and invasion[45,47]
CLC3Regulate cell viability, proliferation, and metastasis[77]
CLC4Regulate invasion, migration, and pH of cytosolic and intracellular organelles[81,82]
CFTRDecreased cell proliferation, migration, and invasion[88]
VDAC1Regulate apoptosis, proliferation, migration, and invasion[98]
AE2Contributed to progression of cancer
Promote tumor cell malignancy
Responsible for elevated Golgi resting pH
[132,137]
Prostate cancerTMEM16ACell proliferation and development to malignant tumor[4]
CLIC1Promoted cancer cell survival, proliferation, and invasion[46]
CLC3Correlation between cell volume regulation and cancer development/ metastasis[70]
CFTRInhibited various cancer-related processes, such as EMT
Enhanced chemo-resistance
[92,94]
Gallbladder cancerCLIC1Enhanced cellular migration and invasion[48,64]
Gastric cancerTMEM16ACell proliferation and development to malignant tumor[7]
CLIC1Enhanced cellular migration and invasion[44,49,61]
AE1Involved in release of p16INK4A and cell growth
Positive correlation with cancer size and metastasis
[126,127,128,129]
AE2Contributed to progression of cancer[133,134]
SLC26A3Interact with AMP18[141]
VRACRegulates proliferation[116]
Brain tumorTMEM16ACell proliferation and development to malignant tumor[8,9]
CLC3Enhanced cell invasion[72]
VDAC2Regulator for the metabolic reprogramming[111]
Clear cell renal cell carcinomaCLIC1Cell invasion[50]
Neuroendocrine tumorCLC3Enhanced resistance to anticancer drug[71]
Esophageal cancerTMEM16ACell proliferation and development to malignant tumor[10]
AE1Related with tumor progression through the crosstalk with MAPK and hedgehog signaling pathways[130]
LiposarcomaTMEM16ACell proliferation and development to malignant tumor[13]
LeiomyosarcomaTMEM16ACell proliferation and development to malignant tumor[14]
Salivary gland cancerTMEM16ACell proliferation and development to malignant tumor[15]
ChondroblastomaTMEM16ACell proliferation and development to malignant tumor[16]
Hydatidiform molesCLIC1Enhanced cellular migration and invasion[43]
Ovarian cancer cellsCLIC3Co-localized with active integrin α5β1
Cell migration and invasion
[67]
CFTREnhanced cell invasion and migration[85]
AE2Involved in tumorigenesis through activation of the mTOR/p70S6K1 pathway[136]
Cervical cancerCFTREnhanced cell invasion and migration[86]
VDAC1Regulate progression of tumorigenesis[105]
Endometrial cancerCFTRInhibited various cancer-related processes, such as EMT[91]
VDAC1Pore-forming role in mitochondrial membrane
Cell growth, proliferation, migration, and invasion
[96]
Oral squamous cell carcinomaBestrophin-1Involved in proliferation[27]
VRACRegulates proliferation
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kim, H.J.; Lee, P.C.-W.; Hong, J.H. Chloride Channels and Transporters: Roles beyond Classical Cellular Homeostatic pH or Ion Balance in Cancers. Cancers 2022, 14, 856. https://doi.org/10.3390/cancers14040856

AMA Style

Kim HJ, Lee PC-W, Hong JH. Chloride Channels and Transporters: Roles beyond Classical Cellular Homeostatic pH or Ion Balance in Cancers. Cancers. 2022; 14(4):856. https://doi.org/10.3390/cancers14040856

Chicago/Turabian Style

Kim, Hyeong Jae, Peter Chang-Whan Lee, and Jeong Hee Hong. 2022. "Chloride Channels and Transporters: Roles beyond Classical Cellular Homeostatic pH or Ion Balance in Cancers" Cancers 14, no. 4: 856. https://doi.org/10.3390/cancers14040856

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

Article Metrics

Back to TopTop