Open Access

Potassium channels as novel molecular targets in hepatocellular carcinoma (Review)

  • Authors:
    • Xingyue Chen
    • Li Zhang
    • Ling He
    • Liming Zheng
    • Biguang Tuo
  • View Affiliations

  • Published online on: August 24, 2023     https://doi.org/10.3892/or.2023.8622
  • Article Number: 185
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Hepatocellular carcinoma (HCC) poses a serious health burden worldwide. It is often not diagnosed until the patient is at an advanced stage of the disease, when treatment options are limited and the prognosis is poor. Therefore, novel treatment strategies are urgently required. Potassium (K+) channels have an important role in HCC, including regulating the proliferation, migration, invasion and drug resistance of HCC cells. The aim of the present review was therefore to survey the relevant publications that have investigated K+ channels not only as markers for the early diagnosis of HCC, but also as potential therapeutic targets for the treatment of HCC. Several of these channels have been indicated to be the sites of action for natural products previously known to inhibit HCC; however, more systematic studies are required to determine which K+ channels may be utilized for the clinical treatment of HCC, particularly in the advanced stages of the disease and in cases where patients are resistant to the existing drugs.

Introduction

Hepatocellular carcinoma (HCC) is a chronic disease and one of the most significant causes of cancer-associated death; furthermore, the clinical prognosis is usually poor (1). The etiology of liver cancer is diverse, including viral infection, alcoholic and non-alcoholic fatty liver disease, aflatoxins, genetic and metabolic factors and infection, among which viral infection [via hepatitis B virus (HBV) and hepatitis C virus (HCV)] is the most important factor (2,3). In recent years, with the prevalence of obesity, diabetes and various metabolic syndromes increasing, the incidence of HCC has also been shown to be on a clear upward trajectory (1,4,5). Liver cancer is divided into primary and secondary HCC, and the most important type of HCC is primary HCC (cancer originating from mutations in hepatocytes or other cell types in the liver), which accounts for ~80% of all cases of HCC; other types of HCC include intrahepatic cholangiocarcinoma and mixed hepatocellular cholangiocarcinoma (3,6). Although various types of liver cancer are significant, the present review will focus mainly on primary HCC. Clinically, the majority of cases of HCC are identified at a nearly advanced stage, in part due to the fact that HCC is not generally associated with obvious physical signs or symptoms in the early stages. By contrast, early HCC can usually only be detected either by ultrasound imaging or by measuring the blood α-fetoprotein concentration, although its specificity is not high (2,6). As a result, HCC is not easily detected in the early stages, resulting in only a small number of, and poor, options of therapy being available. It is worth noting that the treatment of HCC is complex and several years of study have demonstrated that treatment of patients with HCC depends on the clinical manifestation, liver function and tumor staging, although multidisciplinary treatments exist, which mainly include surgical resection and liver transplantation, radiofrequency ablation, chemical drug targeting inhibition and immune suppression (3,7,8). However, due to its high invasiveness and high metastasis rate, the prognosis for patients with HCC continues to be poor (9,10). Therefore, there is an urgent need to identify novel early markers and therapeutic targets to prevent, diagnose and treat the disease.

Ion channels are specialized proteins found in cell membranes, which facilitate the movement of specific ions across the plasma membrane. These are involved in fundamental cellular processes, such as nerve impulse transmission, cell proliferation, apoptosis, hormone secretion and sensory transduction (11,12). The aberrant expression and function of ion channels leads to impairment of these processes, allowing normal cells to transform into malignant derivatives that exhibit uncontrolled proliferation and spread, which are hallmarks of cancer (13). Previous studies have confirmed that ion channels fulfill an important role in the development and progression of cancer, including the infinite proliferation of cells and their invasion and metastasis (1416), and ion channels have been approved as effective drug targets for cancer therapy (17,18). As the most widely distributed type, K+ channels regulate a variety of biological processes by controlling the flow of K+ across cell membranes. The K+ channel family has a total of 78 members, which can be divided into four major groups based on their domains and activation mechanisms: i) Voltage-gated K+ (Kv) channels, ii) calcium-activated K+ (KCa) channels, which themselves are divided into large conductance (BK), medium conductance (IK) and small conductance (SK) channels, iii) inward recirculated K+ channels and iv) two-pore domain K+ channels (15,19) (Fig. 1). As important contributors to the resting membrane potential, K+ channels affect a variety of physiological processes (including regulating the heart rate, muscle contraction, neurotransmitter release, neuronal excitability, cell volume regulation, cell proliferation and differentiation, as well as cell cycle progression, apoptosis and metabolism) through regulating the intracellular K+ concentration (20). There is an increasing body of evidence to suggest that a wide variety of K+ channels are expressed on tumor cells, and dysregulation of their expression has been identified at the genomic, transcriptional, post-translational and epigenetic levels (15). In general, K+ channels have been shown to regulate cell proliferation through four mechanisms: i) Establishment of oscillating membrane potentials; ii) control of cell volume dynamics; iii) regulation of calcium signaling; and iv) promotion of malignant growth through atypical non-ionic osmotic functions (15). Therefore, these channels have been shown to fulfill important roles in regulating tumor cell proliferation, cell cycle progression and apoptosis (21,22) (Fig. 2). For instance, upregulation of Kv1.1 has been demonstrated to be a marker for a subgroup of medulloblastomas (23); elevated levels of Kv1.3 expression are detected in numerous human malignancies, including breast, colon and prostate cancer (24); a high expression level of Kv11.1 [or human ether-a-go-go-related gene (hERG)] was shown to serve as a marker for solid and blood cancers (25); and the overexpression of Kv10.1 [or Ether-a-go-go-1 (EAG1)] has been identified in cancers of various human organs (26). In addition, K+ channel modulators exert antitumor effects primarily as regulators of various types of cancer cell behaviors, including proliferation and migration (27). Furthermore, the abnormal expression and dysfunction of specific K+ channels may lead to the development of various diseases. In addition, the expression and activity of K+ channels have been indicated to be significantly correlated with the grade of malignancy of tumors. Targeting K+ channels may therefore have great therapeutic potential in terms of the treatment of a wide range of human diseases (27,28). Certain studies have indicated that targeting the inhibition of K+ channels may either directly inhibit tumor growth or improve the efficacy of chemotherapy or cytotoxic drugs, and this may be used as a combined treatment strategy for exerting anti-tumor effects (29,30). However, to date, to the best of our knowledge, the role of K+ channels in HCC has rarely been studied.

Therefore, the aim of the present review was to provide an overview of the dysregulation of K+ channels in HCC, also discussing their role in the proliferation, invasion and migration of HCC. The latest progress that has been made in terms of research on drugs targeting K+ channels for the treatment of HCC is also discussed, as well as the existing limitations and future research directions in this field. These research efforts are geared towards providing novel opportunities for the early detection and treatment of HCC.

Role of K+ channels in regulating metabolism, proliferation and injury of liver cells

The liver is a complex organ composed of a variety of different types of cells, which fulfills crucial roles in the body's metabolism. The roles of liver cells mainly comprise carbohydrate, lipid, protein and amino acid metabolism, bile acid intake, synthesis and output, as well as the metabolism of drugs and other foreign substances and the excretion of biological molecules (31). Therefore, damage sustained to the liver cells has an adverse effect on human health. K+ channels fulfill an important role in acute and chronic hepatocyte injury. For instance, margatoxin (MgTX), a Kv1.3-specific blocker, has been shown to reduce the serum levels of TNF-α, IL-6, alanine transaminase and aspartate transaminase to reduce the expression ratio of C-C motif chemokine receptor 2/glutathione reductase 1 double-positive cells and ionized calcium binding adaptor molecule 1/c-type lectin domain family 4 member F positive cells in peripheral mononuclear macrophages, as well as reducing the infiltration rate of peripheral mononuclear macrophages into the liver (32). MgTX has also been shown to markedly protect the liver from acute liver injury. In addition, it is a novel target for the prevention and treatment of alcoholic fatty liver disease, precisely since it is able to regulate the function of macrophages. Further studies have indicated that the Kv1.3 pathway may alleviate the development of liver fibrosis by reducing the expression of TGF-β (32). It has been reported that KCa3.1 expression is increased in hepatocytes with liver fibrosis and that these increases coincide with the progression of liver injury. In addition, the inhibition of KCa3.1 has been shown to lead to cell apoptosis and increase the level of DNA damage, and also to stimulate the proliferation of hepatic stellate cells and aggravate liver fibrosis, which demonstrates that KCa3.1 channels exert a protective role in liver injury (33). The activity of two-pore domain K+ (KCNK2) channels determines resting membrane potential and Ca2+ levels, thereby fulfilling a role in extracellular matrix production and the cell proliferation of hepatic stellate cells, providing a potential therapeutic target for hepatic fibrosis (34).

In recent years, the role of K+ channels in the pathogenesis and treatment of HCC has been gradually uncovered. Several studies have shown that K+ ions are a key regulator of hepatocyte function, which is manifested in inhibiting hepatocyte proliferation and inducing apoptosis, thereby preventing the metastasis of hepatocytes during the process of carcinogenesis. First, K+ has been shown to inhibit the proliferation of hepatocytes, particularly HepG2 cells. In addition, the results of cell-cycle analysis experiments have indicated that K+ is able to block the S-phase of the cell cycle and inhibit the growth of L02 and HepG2 cells via preventing normal DNA replication (35). Furthermore, it is well-established that the pro-apoptotic protein Bax and the anti-apoptotic protein Bcl-2 fulfill a key role in the regulation of cell apoptosis, and that a decrease in the Bcl-2/Bax ratio can promote cell apoptosis. K+ ions promote the expression of the channel protein hERG in a dose-dependent manner, possibly by upregulating the expression of voltage-dependent anion-selective channel protein 1 or through disrupting the balance of the Bcl-2/Bax ratio, which prevents cells from being transferred to the precancerous pathway, leading to the imbalance of the mitochondrial membrane potential, thereby inducing mitochondria to release cytochrome c and to activate caspase proteins (35). These steps consequently result in an imbalance of the caspase-3/7 ratio, eventually leading to apoptosis. In conclusion, targeted activation or inhibition of K+ channels can be a potential therapeutic approach to inhibit the development and progression of HCC.

K+ channels in the development, migration, proliferation and invasion of HCC

Although a large number of ion channels associated with the proliferation, invasion and metastasis of HCC have been reported, the number of specific biomarkers and therapeutic targets available remains limited (36). As one of the most extensive ion channels, various studies have confirmed that K+ channels have an important role in the development, migration, proliferation and invasion of HCC and may be recruitable as novel tumor markers and targeted therapeutic targets (Table I). Although the underlying mechanisms involved have yet to be fully elucidated, this may help to provide a foundation upon which further research can be based.

Table I.

Role of K+ channels in the development of HCC.

Table I.

Role of K+ channels in the development of HCC.

NameRoleTherapeutic strategy(Refs.)
BKExhibits high expression in HCCInhibition(37)
Promotes the migration of HCC cells
KCa3.1Exhibits high expression in HCCInhibition(3840)
Promotes the proliferation, migration and transfer of HCC and ICC cells
EAG1Exhibits high expression in HCCInhibition(41)
Promotes the proliferation, migration and invasion of HCC
KCCN2, KCNK15, KCNK17Low expression in HCC Utility as diagnostic markers and predictors of prognosisRequires further research(42)
KCNK9Exhibits high expression in HCCRequires further(42)
Utility as a diagnostic markerresearch
KCNQ1Shows low expression in HCCActivation(43)
Promotes the invasion of cells
KCNQ1OT1Exhibits high expression in HCCInhibition(4446)
Promotes the proliferation and transfer of HCC cells, and inhibits apoptosis
ATP1A1FXYD6Exhibits high expression in HCCInhibition(4749)
Promotes the occurrence, proliferation and migration of HCC cells
Kv1.3Exhibits high expression in PBC Promotes cell proliferation and apoptosisInhibition(50)

[i] HCC, hepatocellular carcinoma; ICC, intrahepatic cholangiocarcinoma; BK, large-conductance calcium-activated potassium channel; KCa3.1, intermediate-conductance Ca2+-activated K+ channel; EAG1, Ether-a-go-go-1 channel; KCCN2, KCCN15, KCCN17, KCCN9, two-pore domain K+ channels; KCNQ1, potassium voltage-gated channel, KQT-like subfamily, member 1 channel; KCNQ1OT1, long non-coding RNA K+ voltage-gated channel subfamily Q member 1 overlapping transcript 1 channel; ATP1A1FXYD6, Na+/K+-ATPase channel; Kv1.3, voltage-gated potassium channel 1.3.

BK channels

Large-conductance calcium-activated potassium channels, i.e. BK channels, belong to the Ca2+-activated K+ channel family, which also contains two other members, namely IK and SK Ca2+-activated K+ channels (37). BK channels were first identified in chromaffin cells in 1981 (38), and were later found to be expressed in neurons of the vibratory nervous system, where they can be activated by membrane depolarization and increased cytosolic Ca2+ levels (39). Previous studies have reported a variety of functions of BK channels; for instance, BK channels are involved in the regulation of the cell cycle and proliferation, and they have also been shown to be involved in the migration of cancer cells (40). Previous studies have reported on disorders of BK channels in various types of cancer cells, including triple-negative breast cancer cells, neuroblastoma cells, glioblastoma and human astrocytoma cells (40). The patch-clamp technique has also been used to identify BK channels in SMMC-7721 and Huh7 cells. The results obtained indicated that BK channels were functionally expressed in both HCC cells and normal stem cells (40). At the same time, the expression of BK channels in patients with HCC was detected, and it was found that the expression of BK channels in tumor tissues was higher compared with that in non-tumor tissues. More interestingly, the prognosis of patients with a high expression of BK channels was found to be significantly lower compared with that of patients with low BK channel expression (40). A significant role has been identified for BK channels in terms of promoting cancer cell migration and two possible explanations have been proposed: i) Through reducing the expression of epithelial-to-mesenchymal transition (EMT)-associated proteins, such as E-cadherin, vimentin and N-cadherin, and cell-cell contact is thereby reduced, which promotes cancer cell migration; and ii) through reducing cell volume to promote tumor cell migration (40).

KCa3.1 channels

The IK Ca2+-activated K+ channel (KCa3.1) belongs to a medium-conductance calcium-activated K+ channel family and is a potential tumor-targeting molecule, which regulates intracellular ion homeostasis and cell volume under physiological conditions (41). It has been confirmed that this channel is overexpressed in a variety of different types of cancer and regulates the migration, invasion, proliferation and treatment resistance of cancer cells (42,43). It is well-known that HCC stem cells fulfill an important role in tumorigenesis, tumor recurrence and metastasis. A previous study found that KCa3.1 is highly expressed in liver cancer stem cells. KCa3.1 has also been shown to promote the proportions of CD133+ and CD44+ cell subsets, the expression of stem cell transcription factors and the ability of pellet formation in vitro, and to increase the incidence of tumor and tumor growth in vivo (44). Several studies have used immunohistochemical analysis to observe that the expression of KCa3.1 in HCC tissues is significantly upregulated, and this upregulation of gene expression is not only associated with the serum α-fetoprotein level, but it is also associated with an increased risk of recurrence in patients with early HCC (45,46). A number of studies have investigated the potential underlying mechanism(s) governing how KCa3.1 exerts a role in HCC. On the one hand, silencing the expression of KCa3.1 has been shown to lead to a reduction in the levels of extracellular signal-regulated kinases 1 and 2 (ERK1/2) and matrix metalloproteinase 9 (MMP-9) (47). Both ERK1/2 and MMP-9 are considered important biomarkers in the MAPK/ERK signaling pathway. MMP-9 is able to induce the degradation of the extracellular matrix, thereby reducing the stability of cancer cells and making them more prone to metastasis. Activation of ERK1/2 may lead to the transcriptional activation of downstream target genes that are associated with cell proliferation, migration and metastasis (47). In addition, it has been conclusively shown that KCa3.1 promotes the migration of HCC cells through the MAPK/ERK signaling pathway, thereby promoting tumor migration and invasion. Knockdown or inhibition of KCa3.1 expression has also been shown to reduce the migratory and invasive capabilities of HCC cells (46). Alternatively, other studies have found that KCa3.1 can promote the proliferation, invasion and migration of HCC cells via regulating the S-phase kinase-associated protein 2 (SKP2)/P27/P21 signaling pathway. The possible underlying mechanism is that upregulation of KCa3.1 expression changes the half-life of the SKP2 protein, which subsequently mediates the degradation of P21/P27 to promote HCC cell cycle progression. By contrast, knockdown of KCa3.1 was found to significantly reduce the migratory or invasive potential of LM3 and Huh7 cells (45). These results corroborated that KCa3.1 may be a potential molecular target for the treatment of HCC. In HCC, KCa3.1 channels have also been implicated in the development of intrahepatic cholangiocarcinoma (ICC). The expression of the KCa3.1 channel is significant in ICC and this was found to be correlated with the age, lymph node metastasis and TNM stage of the patients. Finally, pharmacological inhibition or knockdown of KCa3.1 was also shown to reduce the proliferative and invasive capabilities of ICC cells (43).

Eag1 channel

The Eag1 channel, also known as Kv10.1, is a voltage-gated K+ channel that is mainly distributed in the cell membrane and participates in various physiological processes of the body, including cell action-potential repolarization, Ca2+ signal transduction and cell volume regulation, thereby promoting cell proliferation and migration (27). It is worth noting that this channel has been found to have carcinogenic properties and that it has therefore garnered great interest among cancer researchers (48). A previous study reported that Eag1 is expressed at a low level in normal tissues, with the exception of the central nervous system and abnormal expression in tumor cells of different origins (26). A previous study also reported that, the mRNA and protein expression of Eag1 in cirrhotic tissues and pretumor lesions was significantly higher compared with that in normal liver (49). To investigate the role of Eag1 in HCC, the authors found that the colony-formation capabilities and proliferation rates of the LM3 and Huh7 cell lines were significantly decreased following downregulation of Eag1. On the other hand, Eag1 overexpression promoted the colony-formation capability and proliferation of the cells. In addition, macroscopic observations of the tumor volume showed that the tumors with Eag1 overexpression were larger compared with those in the control group with normal expression of Eag1 (50). In the same study, the authors identified the putative mechanism to account for the above effects: On the one hand, Eag1 is able to regulate SKP2 through ubiquitination to improve cell proliferation, and the expression of Eag1 was shown to be positively correlated with SKP2. Overexpression of Eag1 led to a prolongation of the half-life of SKP2, which subsequently stimulated the degradation of P21/P27, thereby accelerating the cell cycle progression of HCC. On the other hand, Eag1 has also been shown to promote the migration and invasion of tumor cells via promoting the formation of pseudopodia (50). In conclusion, downregulation of Eag1 expression may be used as a therapeutic strategy for HCC.

KCNK channels

KCNK channels are K+-selective channels. The majority of KCNKs act as outward rectifying channels, or are almost voltage-independent at physiological K+ concentrations to maintain the resting membrane potential, thereby regulating biological metabolism and apoptosis (5153). It is worth noting that KCNKs are able to contribute to oncogenes in various cancers. For instance, in ovarian cancer, certain regulators of KCNK2 have been shown to inhibit apoptosis and to promote cell proliferation (54). The expression level of the long non-coding RNA (lncRNA) KCNK15-antisense 1 was found to be downregulated in pancreatic cancer, which consequently inhibited the invasiveness of tumor cells (55). By contrast, KCNK9 is upregulated in breast and colorectal cancer, and increases tumor tolerance via its anti-apoptotic activity on the cancer cells (56,57). In HCC, different KCNK channels have been demonstrated to have different expression levels. Through analyzing the UALCAN database, researchers observed that the expression levels of KCNK2, KCNK15 and KCNK17 were decreased in HCC, whereas the expression level of KCNK9 was increased, and these trends were associated with poor prognosis for patients with HCC. Additionally, the receiver operating characteristic curve analysis suggested that the levels of KCNK2, KCNK9, KCNK15 and KCNK17 levels may be used as biomarkers for the diagnosis and prognosis of HCC (52). This study provided important information for the early diagnosis of HCC.

Potassium voltage-gated channel, KQT-like subfamily, member 1 (KCNQ1) channels

KCNQ1 channels, as a class of voltage-gated K+ channels, are usually expressed in a variety of tissues, including the heart, stomach, intestine and pancreas, which mediate the outflow of K+ ions from cells and regulate ion homeostasis in tissues (58). Previous studies have confirmed that the expression of KCNQ1 is markedly downregulated in human HCC cell lines compared with non-malignant cells or normal human liver tissues (59,60). On the other hand, overexpression of KCNQ1 inhibited the invasion of HCC, and the same study revealed that pharmacological activation of KCNQ1 channels inhibited tumor metastasis of HCC in nude mice. The putative underlying mechanism may be that KCNQ1 affects the cellular distribution of β-catenin, thereby inhibiting the activity of the Wnt/β-catenin signaling pathway, which acts as one of the main pathways involved in both the initiation and progression of HCC (59) and in the mRNA expression of its downstream targets, ultimately fulfilling a tumor-suppressor role (60). In conclusion, it has been demonstrated that the KCNQ1 channel may be used as both a biomarker and potential therapeutic target for HCC.

Long non-coding RNA (lncRNA) K+ voltage-gated channel subfamily Q member 1 overlapping transcript 1 (KCNQ1OT1) channels

The lncRNA KCNQ1OT1 is a chromatin-regulatory lncRNA that has been shown to participate in the regulation of various types of cancer as an oncogene, including rectal cancer and lung cancer (61). However, at present, little is known concerning the mechanism via which KCNQ1OT1 promotes carcinogenesis. However, certain studies have found that the short-strand repeat polymorphism in KCNQ1OT1 contributes to the initiation of HCC, which may affect the expression of KCNQ1OT1 and cyclin-dependent kinase inhibitor 1C (CDKN1C) through a structure-dependent mechanism (62). Previous studies have also reported that the expression of KCNQ1OT1 is associated with the development of HCC. Several studies have reported that KCNQ1OT1 is able to affect the growth of HCC; for example, by inhibiting the expression of microRNA (miR-504) (61), through targeting miR-338-3p (63), by regulating the miR-506-3p/forkhead box (Fox)Q1 axis (64) and through regulating the miR-146A-5p/alkaline ceramidase 3 signaling axis (65). Of note, the latter study (65) reported that the expression of KCNQ1OT1 is significantly upregulated in HCC; furthermore, it was found that silencing KCNQ1OT1 inhibited cell proliferation, improved the sensitivity to radiotherapy and promoted cell apoptosis, as well as hindering the metastasis of HCC cells. In terms of treatment strategies, a previous study (66) reported that KcNQ1OT1 knockdown enhanced the sensitivity of sorafenib through targeting miR-506, induce cell apoptosis and inhibit the metastasis of sorafenib-resistant HCC cells. In conclusion, KCNQ1OT1 may also provide novel therapeutic strategies and opportunities for HCC.

Na+/K+-ATPase (NKA) channel

NKA channel, as a member of the P-type ATPase family, is composed of three peptides: The α and β subunits and FXYD protein (a type of small molecule single transmembrane protein, which regulators of Na+/K+-ATPase). It pumps three Na+ ions out and two K+ ions in for each ATPase hydrolyzed, acting as a multifunctional protein, which fulfills roles in cell attachment, adhesion, motility and signal transduction (67,68). In recent years, it was shown that NKA is abnormally expressed and has abnormal activity in various types of cancer, which signified that it may be anticipated to become a novel target for tumor therapy. Of note, the dysregulation of NKA subunits has been shown to vary among different types of cancer. Previous studies have found that the expression of ATPase Na+/K+ transporting subunit α1 (ATP1A1) in HCC is significantly higher compared with that in adjacent non-tumor tissues (69,70). ATP1A1 has been shown to have the following roles: i) Knockdown of ATP1A1 in HepG2 and MHCC97H cells significantly reduced their proliferation in vitro and inhibited the tumorigenicity of MHCC97H cells in vivo; ii) downregulation of ATP1A1 expression in of HepG2 cells led to cell-cycle arrest in G2/M phase and apoptosis, and in Hep3B, it increased cell migration; and iii) downregulation of ATP1A1 expression led to the production of excessive reactive oxygen species (ROS), which led to DNA damage and cell-cycle arrest, and prevented the replication of damaged and defective DNA (69). In addition, in non-alcoholic steatohepatitis (NASH)-associated malignancies, α1-NKA signaling was shown to activate the PI3K/Akt signaling pathway, which simultaneously inhibited the FoxO3 circuit, leading to the downregulation of the anti-apoptotic survival protein and pro-apoptotic protein, second mitochondria-derived activator of caspase/direct inhibitor of apoptosis-binding protein with low pI, a process that is conducive to cell division and the development of HCC (71). Therefore, ATP1A1 not only serves as a potential biomarker for the diagnosis and prognosis of HCC (72), but it may also offer a therapeutic path for HCC. FXYD proteins are regulators of Na+/K+-ATPase that are located in the cell membrane, which regulate the kinetic properties of Na+/K+-ATPase by changing the rate and affinity of Na+ and K+ transport (73). FXYD6 was shown to be upregulated in HCC, promoting the migration and proliferation of HCC cells (74). The upregulation of FXYD6 is also positively correlated with an increase of Na+/K+-ATPase activity and it mainly exerts its antitumor activity through activating the downstream SrC-ERK signaling pathway. In addition, the blocking of FXYD6 by self-produced anti-FXYD6 functional antibody was shown to significantly inhibit the growth of mouse xenograft tumors, indicating that FXYD6 can act as a novel therapeutic target for HCC (74). Taken together, these results demonstrated that FXYD6 fulfills a key role in the progression of HCC, suggesting that FXYD6-targeting therapy may be of benefit for the clinical treatment of patients with HCC. Therefore, each subunit of NKA may be used as a novel potential therapeutic target for HCC, offering further options for the treatment of HCC.

Kv1.3

Kv1.3, an important regulatory protein of the immune response, has been found in lymphocytes and is mainly involved in immune regulation of the body's immune system (75). Kv1.3 protein on cell membranes is involved in cell proliferation, whereas Kv1.3 channels located on mitochondria are involved in cell apoptosis, and therefore, Kv1.3 located on mitochondria is considered to be a novel tumor biomarker (32,76). It has been reported that the Kv1.3 channel is involved in the proliferation and apoptosis of a variety of different types of tumor. It is noteworthy that the expression of Kv1.3 has been shown to vary with the tumor stage and downregulation of Kv1.3 may significantly inhibit cell proliferation and increase cell apoptosis (77). However, to date, only a small number of studies have been published on the role of this channel in HCC, although a previous study reported that Kv1.3 blocker can regulate the hyperreactivity of B lymphocytes and inhibit the abnormal hypersecretion of antimitochondrial antibodies in primary biliary cirrhosis to achieve the purpose of treating HCC (78). However, the precise role of Kv1.3 in HCC requires further study.

Targeted therapeutic agents for HCC

At present, the treatment of HCC mainly includes surgery, radiotherapy and chemotherapy, although HCC is not sensitive to radiotherapy and conventional chemotherapy drugs, such as doxorubicin, fluorouracil and cisplatin, have serious side effects (7,79). Sorafenib is the first-line drug in the treatment of HCC supported by the currently available data, although the efficacy varies among different patients and the sensitivity to the drug is typically reduced following long-term treatment (80); therefore, it is urgent to identify novel therapeutic strategies. With this as the aim, molecular targeted therapy has been attracting increasing attention. Research confirming that the abnormal expression of certain K+ channels serves an important role in the development and progression of HCC, has also found several drugs able to target and inhibit the ion channels, thereby exerting antitumor effects (Table II). For instance, pharmacological inhibition of voltage-gated K+ channels has long been reported to reduce the adhesion and proliferation of HCC cells, thereby exerting their antitumor effects (81).

Table II.

Drugs targeting K+ channels in HCC.

Table II.

Drugs targeting K+ channels in HCC.

DrugsMechanism(Refs.)
IbTXBlocks the BK channel(37)
Inhibits migration and invasion
AstemizoleBlocks the Eag1 channel(82)
Inhibits proliferation and promotes apoptosis
Procyanidin B1Blocks the Eag1 channel(79)
Inhibits proliferation and promotes apoptosis
TRAM-34Blocks the KCa3.1 channel(38,83)
Inhibits proliferation and promotes apoptosis
Berberine and OuabainCombined with Na+/K+-ATPase enhances the anti-liver cancer effect of sorafenib(47,84)
Sodium orthovanadateInhibition of ATPase reverses sorafenib resistance of HCC cells(85)

[i] HCC, hepatocellular carcinoma; Eag1, Ether-a-go-go-1; IbTX, iberiotoxin; TRAM-34, 1-[(2-chlorophenyl) diphenylmethyl]-1H-pyrazole.

Iberiotoxin (IbTX)

IbTX, a BK channel antagonist, selectively binds to the pore-forming α-subunit of the BK channel (40). It has been reported that blockade of BK channels inhibits both hypoxia-induced migration and chemotherapy resistance to cisplatin in human glioblastoma cells (82). In HCC, the authors of a previously published study (40) experimentally found that blocking BK channels with IbTX led to a marked inhibition of the migration and invasion of HCC cells under hypoxic conditions (40). Subsequently, the same authors identified that the mechanism of its action was to induce G2 phase arrest of HCC cells, thereby inhibiting their migration and invasion, and regulating the growth of the tumor (40). This finding provided a novel strategy for the treatment of HCC.

Astemizole

Astemizole is an antihistamine that penetrates lipid bilayers and binds to channels inside cell membranes (83). Owing to its inhibitory effects on several cancer-associated proteins, including K+ channels, histamine receptors and P-glycoproteins, research has focused on its potential therapeutic role in cancer. A previous study identified that astemizole was able to inhibit the proliferation and increase the apoptosis of HepG2 and Huh-7 cells (84). Although astemizole has diverse targets, exploration of the mRNA and protein expression levels of Eag1 in these cells has revealed that the inhibition of Eag1 channels mediated by astemizole in HCC may potentially be the mechanism that accounts for its anti-proliferative and pro-apoptotic effects (49,85). Treatment with astemizole was also shown to reduce the mRNA and protein expression levels of Eag1 in diethylnitrosamine-treated mice, resulting in both improved histological features and appearance of the liver (85). It was thereby determined that astemizole may have clinical utility in terms of the prevention and treatment of HCC. Another study reported that serious adverse reactions to the use of astemizole as a therapeutic agent only occur in excessive use of the drug with HCC (84), and thus, it holds promise as a selective agent both to prevent the risk of HCC and as a promising therapeutic agent for patients with HCC.

Procyanidin B1

Procyanidin B1, a natural compound derived from grape seed, not only induces the apoptosis of cells, but also inhibits tumor growth (86,87). A previous study reported that procyanidin B1 directly binds to the Eag1 channel, thereby inhibiting its current, whereas its effect on other K+ channels was negligible, and this provided the putative underlying mechanism to account for its inhibition of the migration and proliferation of HCC cells (79). Of note, procyanidin B1 was found to not exert any adverse effects on normal metabolism in mice compared with conventional antitumor drugs. In conclusion, procyanidin B1 was demonstrated to be a significant potential antitumor drug for HCC.

1-[(2-Chlorophenyl)diphenylmethyl]-1H-pyrazole (TRAM-34)

A previous study reported that inhibition of KCa3.1 via genetic and pharmacological means led to a significant reduction in the proliferation of tumor cells and the susceptibility of tumors to certain therapeutic interventions was also changed (88). TRAM-34, a specific KCa3.1 blocker, has been reported to be effective in inhibiting cell proliferation and motility in a variety of different types of cancer, including glioblastoma and lung cancer (89,90). Of note, long-term treatment with TRAM-34 for atherosclerosis therapy at therapeutic concentrations did not lead to any significant side effects (91). TRAM-34 has also been shown to inhibit the proliferation and induce the apoptosis of HepG2 cells. On the one hand, TRAM-34 can inhibit cell proliferation by mediating decreases in both the mRNA expression of estrogen receptor-α and nuclear factor-κB activation (92); on the other hand, the apoptosis of HCC cells was found to be promoted by regulating the intracellular level of ROS and through promoting p53 activation. In addition, treatment with TRAM-34 also led to a marked inhibition of the migration of HepG2, thereby reducing the development of HCC (93). In addition to fulfilling a role in HCC, inhibition of KCa3.1 channels by TRAM-34 was also shown to inhibit hepatocyte fibrosis (94) and ICC tumor growth (43). Taken together, these results indicated that TRAM-34 may potentially be a drug for the treatment of HCC.

Berberine and ouabain

Berberine, a natural dibenzyl isoquinoline alkaloid isolated from Berberis (common name: Barberry), has been studied as a drug against a variety of different types of cancer, including HCC. It was demonstrated that berberine could induce the phosphorylation of Src in a Na+/K+-ATPase-dependent manner, leading to activation of p38-MAPK and the epidermal growth factor receptor (EGFR)/ERK signaling pathway (95). In addition, the Na+/K+-ATPase ligand ouabain has also been shown to induce the phosphorylation of Src, EGFR, insulin-like growth factor 1 receptor, ERK1/2 and p38-MAPK in HCC cells, leading to the inhibition of cell growth and migration through inhibiting EMT in HCC cells both in vivo and in vitro (69). It was found that treatment of HCC with sorafenib led to a significant induction of cell death and inhibition of the growth of HCC xenografts in vivo (95). Therefore, targeting the Na+/K+-ATPase with berberine and ouabain is a novel strategy to enhance the effects of sorafenib. In addition to the drugs that have already been studied, it would be useful to identify other drugs that target this channel in future studies.

Sodium orthovanadate (SOV)

It has been reported that Na+/K+-ATPase activity is increased in drug-resistant tumors and its enhanced activity contributes towards the biological behaviors observed in, and drug resistance of, cancer, such as prostate, breast and lung cancer or leukemia (96). Blocking Na+/K+-ATPase through the use of specific inhibitors has been shown to re-sensitize cancer to chemotherapy drugs (97,98). It was found that the sorafenib resistance of HCC cells was associated with higher levels of Na+/K+-ATPase activity. SOV, a phosphate analogue, is a recognized ATPase inhibitor and treatment of sorafenib-resistant HCC with SOV has been shown to re-sensitize the cancer to sorafenib, enhancing its antitumor effects (99). Although the exact mechanism underlying its inhibition of ATPase requires further study, what has been discovered to date is at least sufficient to demonstrate that SOV may be an effective candidate drug to overcome sorafenib resistance in the treatment of HCC.

Conclusions and prospects

A large number of studies have demonstrated that the regulation of ion channels is associated with the development and progression of HCC (77). In the present review, the abnormal expression of K+ channels in HCC was discussed. These K+ channels are known to be involved in the development, proliferation and invasion of HCC, and so they may serve as novel tumor markers and potential therapeutic targets for HCC. Although an increasing body of evidence has indicated the abnormal expression and function of K+ channels in HCC, research on ion-channel-targeted therapy in cancer remains in its infancy and the mechanisms of action have yet to be fully elucidated. Therefore, further systematic exploration of the mechanisms involved is required to help improve the quality of life of patients with HCC. Although certain K+ channels have been shown to be abnormally expressed in HCC, whether they can be selectively targeted in tumor cells remains to be determined. Although the expression levels of K+ channels in tumor and non-tumor tissues have been compared, the differences in K+ channel expression at the different stages of HCC remain poorly understood. The majority of channels studied so far have been confined to the plasma membrane, but these ion channels may also fulfill important roles in other organelles, such as the mitochondria, and these may be useful as novel targets for tumor therapy in the future. The majority of studies performed to date have been laboratory-based and further studies are required. In the future, it is expected that these targeted drugs will be tested in clinical trials and their efficacy either alone or in combination with other antitumor drugs in patients with HCC or at risk of developing HCC may be further tested. To date, differences between K+ channels in HCC and other tumors have not been reported, and whether K+ channels may be used to distinguish different cancers by comparing the expression levels or expression sites requires further research.

Acknowledgements

Not applicable.

Funding

The present study was funded by the National Natural Science Foundation of China (grant no. 82073087), and the Collaborative Innovation Center of Chinese Ministry of Education (grant no. 2020-39).

Availability of data and materials

Not applicable.

Authors' contributions

XC made substantial contributions to the conception and design of the study, as well as writing the manuscript and performing the literature search. LZha, LHe, LZhe and BT were involved in revising the manuscript critically for important intellectual content. Data authentication is not applicable. All authors have read and approved the final version of the 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 

McGlynn KA, Petrick JL and El-Serag HB: Epidemiology of hepatocellular carcinoma. Hepatology. 73 (Suppl 1):S4–S13. 2021. View Article : Google Scholar

2 

Fujiwara N, Friedman S, Goossens N and Hoshida Y: Risk factors and prevention of hepatocellular carcinoma in the era of precision medicine. J Hepatol. 68:526–549. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Villanueva A: Hepatocellular carcinoma. N Engl J Med. 380:1450–1462. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Calderaro J, Ziol M, Paradis V and Zucman-Rossi J: Molecular and histological correlations in liver cancer. J Hepatol. 71:616–630. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Craig A, von Felden J, Garcia-Lezana T, Sarcognato S and Villanueva A: Tumour evolution in hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 17:139–152. 2020. View Article : Google Scholar : PubMed/NCBI

6 

Sia D, Villanueva A, Friedman S and Llovet J: Liver cancer cell of origin, molecular class, and effects on patient prognosis. Gastroenterology. 152:745–761. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Yang JD, Hainaut P, Gores GJ, Amadou A, Plymoth A and Roberts LR: A global view of hepatocellular carcinoma: Trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol. 16:589–604. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Bruix J, Gores GJ and Mazzaferro V: Hepatocellular carcinoma: Clinical frontiers and perspectives. Gut. 63:844–855. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Llovet J, Montal R, Sia D and Finn R: Molecular therapies and precision medicine for hepatocellular carcinoma. Nat Rev Clin Oncol. 15:599–616. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Chan LK and Ng IO: Joining the dots for better liver cancer treatment. Nat Rev Gastroenterol Hepatol. 17:74–75. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Kunzelmann K: Ion channels and cancer. J Membr Biol. 205:159–173. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Lang F, Föller M, Lang KS, Lang PA, Ritter M, Gulbins E, Vereninov A and Huber SM: Ion channels in cell proliferation and apoptotic cell death. J Membr Biol. 205:147–157. 2005. View Article : Google Scholar : PubMed/NCBI

13 

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

14 

Prevarskaya N, Skryma R and Shuba Y: Ion channels and the hallmarks of cancer. Trends Mol Med. 16:107–121. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Huang X and Jan LY: Targeting potassium channels in cancer. J Cell Biol. 206:151–162. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Conti M: Targeting K+ channels for cancer therapy. J Exp Ther Oncol. 4:161–166. 2004.PubMed/NCBI

17 

Teisseyre A, Gąsiorowska J and Michalak K: Voltage-gated potassium channels Kv1.3-potentially new molecular target in cancer diagnostics and therapy. Adv Clin Exp Med. 24:517–524. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Kale VP, Amin SG and Pandey MK: Targeting ion channels for cancer therapy by repurposing the approved drugs. Biochim Biophys Acta. 1848:2747–2755. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Kuang Q, Purhonen P and Hebert H: Structure of potassium channels. Cell Mol Life Sci. 72:3677–3693. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Bates E: Ion channels in development and cancer. Annu Rev Cell Dev Biol. 31:231–247. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Comes N, Serrano-Albarrás A, Capera J, Serrano-Novillo C, Condom E, Ramón Y Cajal S, Ferreres JC and Felipe A: Involvement of potassium channels in the progression of cancer to a more malignant phenotype. Biochim Biophys Acta. 1848:2477–2492. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Zúñiga L, Cayo A, Gonzalez W, Vilos C and Zúñiga R: Potassium channels as a target for cancer therapy: Current perspectives. Onco Targets Ther. 15:783–797. 2022. View Article : Google Scholar : PubMed/NCBI

23 

Taylor MD, Northcott PA, Korshunov A, Remke M, Cho YJ, Clifford SC, Eberhart CG, Parsons DW, Rutkowski S, Gajjar A, et al: Molecular subgroups of medulloblastoma: The current consensus. Acta Neuropathol. 123:465–472. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Comes N, Bielanska J, Vallejo-Gracia A, Serrano-Albarrás A, Marruecos L, Gómez D, Soler C, Condom E, Ramón Y Cajal S, Hernández-Losa J, et al: The voltage-dependent K(+) channels Kv1.3 and Kv1.5 in human cancer. Front Physiol. 4:2832013. View Article : Google Scholar : PubMed/NCBI

25 

Pillozzi S, Masselli M, De Lorenzo E, Accordi B, Cilia E, Crociani O, Amedei A, Veltroni M, D'Amico M, Basso G, et al: Chemotherapy resistance in acute lymphoblastic leukemia requires hERG1 channels and is overcome by hERG1 blockers. Blood. 117:902–914. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Hemmerlein B, Weseloh RM, Mello de Queiroz F, Knötgen H, Sánchez A, Rubio ME, Martin S, Schliephacke T, Jenke M, Heinz-Joachim-Radzun, et al: Overexpression of Eag1 potassium channels in clinical tumours. Mol Cancer. 5:412006. View Article : Google Scholar : PubMed/NCBI

27 

Pardo LA and Stühmer W: The roles of K(+) channels in cancer. Nat Rev Cancer. 14:39–48. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Bachmann M, Li W, Edwards MJ, Ahmad SA, Patel S, Szabo I and Gulbins E: Voltage-gated potassium channels as regulators of cell death. Front Cell Dev Biol. 8:6118532020. View Article : Google Scholar : PubMed/NCBI

29 

Shen Z, Yang Q and You Q: Researches toward potassium channels on tumor progressions. Curr Top Med Chem. 9:322–329. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Wang Z: Roles of K+ channels in regulating tumour cell proliferation and apoptosis. Pflugers Arch. 448:274–286. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Ben-Moshe S and Itzkovitz S: Spatial heterogeneity in the mammalian liver. Nat Rev Gastroenterol Hepatol. 16:395–410. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Liu J, Lv XW, Zhang L, Wang H, Li J and Wu B: Review on biological characteristics of Kv1.3 and its role in liver diseases. Front Pharmacol. 12:6525082021. View Article : Google Scholar : PubMed/NCBI

33 

Sevelsted Møller L, Fialla AD, Schierwagen R, Biagini M, Liedtke C, Laleman W, Klein S, Reul W, Koch Hansen L, Rabjerg M, et al: The calcium-activated potassium channel KCa3.1 is an important modulator of hepatic injury. Sci Rep. 6:287702016. View Article : Google Scholar : PubMed/NCBI

34 

Kondo R, Deguchi A, Kawata N, Suzuki Y and Yamamura H: Involvement of TREK1 channels in the proliferation of human hepatic stellate LX-2 cells. J Pharmacol Sci. 148:286–294. 2022. View Article : Google Scholar : PubMed/NCBI

35 

Xia Z, Huang X, Chen K, Wang H, Xiao J, He K, Huang R, Duan X, Liu H, Zhang J and Xiang G: Proapoptotic role of potassium ions in liver cells. Biomed Res Int. 2016:17291352016. View Article : Google Scholar : PubMed/NCBI

36 

Craig A and Villanueva A: Liver capsule: Molecular-based signatures in hepatocellular carcinoma. Hepatology. 63:20182016. View Article : Google Scholar : PubMed/NCBI

37 

Ghatta S, Nimmagadda D, Xu X and O'Rourke ST: Large-conductance, calcium-activated potassium channels: Structural and functional implications. Pharmacol Ther. 110:103–116. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Marty A: Ca-dependent K channels with large unitary conductance in chromaffin cell membranes. Nature. 291:497–500. 1981. View Article : Google Scholar : PubMed/NCBI

39 

Knaus HG, Schwarzer C, Koch RO, Eberhart A, Kaczorowski GJ, Glossmann H, Wunder F, Pongs O, Garcia ML and Sperk G: Distribution of high-conductance Ca(2+)-activated K+ channels in rat brain: Targeting to axons and nerve terminals. J Neurosci. 16:955–963. 1996. View Article : Google Scholar : PubMed/NCBI

40 

He Y, Lin Y and He F, Shao L, Ma W and He F: Role for calcium-activated potassium channels (BK) in migration control of human hepatocellular carcinoma cells. J Cell Mol Med. 25:9685–9696. 2021. View Article : Google Scholar : PubMed/NCBI

41 

Wulff H and Castle N: Therapeutic potential of KCa3.1 blockers: Recent advances and promising trends. Expert Rev Clin Pharmacol. 3:385–396. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Todesca LM, Maskri S, Brömmel K, Thale I, Wünsch B, Koch O and Schwab A: Targeting Kca3.1 channels in cancer. Cell Physiol Biochem. 55:131–144. 2021. View Article : Google Scholar : PubMed/NCBI

43 

Song P, Du Y, Song W, Chen H, Xuan Z, Zhao L, Chen J, Chen J, Guo D, Jin C, et al: KCa3.1 as an effective target for inhibition of growth and progression of intrahepatic cholangiocarcinoma. J Cancer. 8:1568–1578. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Fan J, Tian R, Yang X, Wang H, Shi Y, Fan X, Zhang J, Chen Y, Zhang K, Chen Z and Li L: KCNN4 promotes the stemness potentials of liver cancer stem cells by enhancing glucose metabolism. Int J Mol Sci. 23:69582022. View Article : Google Scholar : PubMed/NCBI

45 

Du Y, Song W, Chen J, Chen H, Xuan Z, Zhao L, Chen J, Jin C, Zhou M, Tuo B, et al: The potassium channel KCa3.1 promotes cell proliferation by activating SKP2 and metastasis through the EMT pathway in hepatocellular carcinoma. Int J Cancer. 145:503–516. 2019. View Article : Google Scholar : PubMed/NCBI

46 

Li QT, Feng YM, Ke ZH, Qiu MJ, He XX, Wang MM, Li YN, Xu J, Shi LL and Xiong ZF: KCNN4 promotes invasion and metastasis through the MAPK/ERK pathway in hepatocellular carcinoma. J Investig Med. 68:68–74. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Ranjan A, Iyer SV, Ward C, Link T, Diaz FJ, Dhar A, Tawfik OW, Weinman SA, Azuma Y, Izumi T and Iwakuma T: MTBP inhibits the Erk1/2-Elk-1 signaling in hepatocellular carcinoma. Oncotarget. 9:21429–21443. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Rodríguez-Rasgado J, Acuña-Macías I and Camacho J: Eag1 channels as potential cancer biomarkers. Sensors (Basel). 12:5986–5995. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Chávez-López MG, Zúñiga-García V, Pérez-Carreón JI, Avalos-Fuentes A, Escobar Y and Camacho J: Eag1 channels as potential early-stage biomarkers of hepatocellular carcinoma. Biologics. 10:139–148. 2016.PubMed/NCBI

50 

Chen J, Xuan Z, Song W, Han W, Chen H, Du Y, Xie H, Zhao Y, Zheng S and Song P: EAG1 enhances hepatocellular carcinoma proliferation by modulating SKP2 and metastasis through pseudopod formation. Oncogene. 40:163–176. 2021. View Article : Google Scholar : PubMed/NCBI

51 

Lotshaw D: Biophysical, pharmacological, and functional characteristics of cloned and native mammalian two-pore domain K+ channels. Cell Biochem Biophys. 47:209–256. 2007. View Article : Google Scholar : PubMed/NCBI

52 

Kheradpezhouh E, Ma L, Morphett A, Barritt GJ and Rychkov GY: TRPM2 channels mediate acetaminophen-induced liver damage. Proc Natl Acad Sci USA. 111:3176–3181. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Li WC, Xiong ZY, Huang PZ, Liao YJ, Li QX, Yao ZC, Liao YD, Xu SL, Zhou H, Wang QL, et al: KCNK levels are prognostic and diagnostic markers for hepatocellular carcinoma. Aging (Albany NY). 11:8169–8182. 2019. View Article : Google Scholar : PubMed/NCBI

54 

Innamaa A, Jackson L, Asher V, van Schalkwyk G, Warren A, Keightley A, Hay D, Bali A, Sowter H and Khan R: Expression and effects of modulation of the K2P potassium channels TREK-1 (KCNK2) and TREK-2 (KCNK10) in the normal human ovary and epithelial ovarian cancer. Clin Transl Oncol. 15:910–918. 2013. View Article : Google Scholar : PubMed/NCBI

55 

He Y, Hu H, Wang Y, Yuan H, Lu Z, Wu P, Liu D, Tian L, Yin J, Jiang K and Miao Y: ALKBH5 inhibits pancreatic cancer motility by decreasing long non-coding RNA KCNK15-AS1 methylation. Cell Physiol Biochem. 48:838–846. 2018. View Article : Google Scholar : PubMed/NCBI

56 

Alvarez-Baron C, Jonsson P, Thomas C, Dryer S and Williams C: The two-pore domain potassium channel KCNK5: Induction by estrogen receptor alpha and role in proliferation of breast cancer cells. Mol Endocrinol. 25:1326–1336. 2011. View Article : Google Scholar : PubMed/NCBI

57 

Kim CJ, Cho YG, Jeong SW, Kim YS, Kim SY, Nam SW, Lee SH, Yoo NJ, Lee JY and Park WS: Altered expression of KCNK9 in colorectal cancers. APMIS. 112:588–594. 2004. View Article : Google Scholar : PubMed/NCBI

58 

Peroz D, Rodriguez N, Choveau F, Baró I, Mérot J and Loussouarn G: Kv7.1 (KCNQ1) properties and channelopathies. J Physiol. 586:1785–1789. 2008. View Article : Google Scholar : PubMed/NCBI

59 

White BD, Chien AJ and Dawson DW: Dysregulation of Wnt/β-catenin signaling in gastrointestinal cancers. Gastroenterology. 142:219–232. 2012. View Article : Google Scholar : PubMed/NCBI

60 

Fan H, Zhang M and Liu W: Hypermethylated KCNQ1 acts as a tumor suppressor in hepatocellular carcinoma. Biochem Biophys Res Commun. 503:3100–3107. 2018. View Article : Google Scholar : PubMed/NCBI

61 

Li C, Miao R, Zhang J, Qu K and Liu C: Long non-coding RNA KCNQ1OT1 mediates the growth of hepatocellular carcinoma by functioning as a competing endogenous RNA of miR-504. Int J Oncol. 52:1603–1612. 2018.PubMed/NCBI

62 

Wan J, Huang M, Zhao H, Wang C, Zhao X, Jiang X, Bian S, He Y and Gao Y: A novel tetranucleotide repeat polymorphism within KCNQ1OT1 confers risk for hepatocellular carcinoma. DNA Cell Biol. 32:628–634. 2013. View Article : Google Scholar : PubMed/NCBI

63 

Zhong W, Dai Q and Huang Q: Effect of lncRNA KCNQ1OT1 on autophagy and drug resistance of hepatocellular carcinoma cells by targeting miR-338-3p. Cell Mol Biol (Noisy-le-grand). 66:191–196. 2020. View Article : Google Scholar : PubMed/NCBI

64 

Jiang M, Cui BW, Wu YL, Zhang Y, Shang Y, Liu J, Yang HX, Qiao CY, Zhan ZY, Ye H, et al: P2X7R orchestrates the progression of murine hepatic fibrosis by making a feedback loop from macrophage to hepatic stellate cells. Toxicol Lett. 333:22–32. 2020. View Article : Google Scholar : PubMed/NCBI

65 

Yang G, Zhou L, Xu Q, Meng F, Wan Y, Meng X, Wang L and Zhang L: LncRNA KCNQ1OT1 inhibits the radiosensitivity and promotes the tumorigenesis of hepatocellular carcinoma via the miR-146a-5p/ACER3 axis. Cell Cycle. 19:2519–2529. 2020. View Article : Google Scholar : PubMed/NCBI

66 

Zhang J, Zhao X, Ma X, Yuan Z and Hu M: KCNQ1OT1 contributes to sorafenib resistance and programmed death-ligand-1-mediated immune escape via sponging miR-506 in hepatocellular carcinoma cells. Int J Mol Med. 46:1794–1804. 2020.PubMed/NCBI

67 

Xie Z and Askari A: Na(+)/K(+)-ATPase as a signal transducer. Eur J Biochem. 269:2434–2439. 2002. View Article : Google Scholar : PubMed/NCBI

68 

Rajasekaran SA, Palmer LG, Quan K, Harper JF, Ball WJ Jr, Bander NH, Peralta Soler A and Rajasekaran AK: Na,K-ATPase beta-subunit is required for epithelial polarization, suppression of invasion, and cell motility. Mol Biol Cell. 12:279–295. 2001. View Article : Google Scholar : PubMed/NCBI

69 

Zhuang L, Xu L, Wang P, Jiang Y, Yong P, Zhang C, Zhang H, Meng Z and Yang P: Na+/K+-ATPase α1 subunit, a novel therapeutic target for hepatocellular carcinoma. Oncotarget. 6:28183–28193. 2015. View Article : Google Scholar : PubMed/NCBI

70 

Xu ZW, Wang FM, Gao MJ, Chen XY, Hu WL and Xu RC: Targeting the Na(+)/K(+)-ATPase alpha1 subunit of hepatoma HepG2 cell line to induce apoptosis and cell cycle arresting. Biol Pharm Bull. 33:743–751. 2010. View Article : Google Scholar : PubMed/NCBI

71 

Udoh US, Banerjee M, Rajan PK, Sanabria JD, Smith G, Schade M, Sanabria JA, Nakafuku Y, Sodhi K, Pierre SV, et al: Tumor-suppressor role of the α1-Na/K-ATPase signalosome in NASH related hepatocellular carcinoma. Int J Mol Sci. 23:73592022. View Article : Google Scholar : PubMed/NCBI

72 

Tang S, Yang X, Zhou C, Mei Y, Ye J, Zhang X, Feng G, Zhang W, Zhang X and Fan W: Sodium pump Na + /K + ATPase subunit α1-targeted positron emission tomography imaging of hepatocellular carcinoma in mouse models. Mol Imaging Biol. 24:384–393. 2022. View Article : Google Scholar : PubMed/NCBI

73 

Garty H and Karlish SJD: Role of FXYD proteins in ion transport. Annu Rev Physiol. 68:431–459. 2006. View Article : Google Scholar : PubMed/NCBI

74 

Gao Q, Chen X, Duan H, Wang Z, Feng J, Yang D, Song L, Zhou N and Yan X: FXYD6: A novel therapeutic target toward hepatocellular carcinoma. Protein Cell. 5:532–543. 2014. View Article : Google Scholar : PubMed/NCBI

75 

Feske S, Wulff H and Skolnik EY: Ion channels in innate and adaptive immunity. Annu Rev Immunol. 33:291–353. 2015. View Article : Google Scholar : PubMed/NCBI

76 

Teisseyre A, Palko-Labuz A, Sroda-Pomianek K and Michalak K: Voltage-gated potassium channel Kv1.3 as a target in therapy of cancer. Front Oncol. 9:9332019. View Article : Google Scholar : PubMed/NCBI

77 

Zúñiga-García V, Chávez-López Mde G, Quintanar-Jurado V, Gabiño-López NB, Hernández-Gallegos E, Soriano-Rosas J, Pérez-Carreón JI and Camacho J: Differential expression of ion channels and transporters during hepatocellular carcinoma development. Dig Dis Sci. 60:2373–2383. 2015. View Article : Google Scholar : PubMed/NCBI

78 

Prosdocimi E, Checchetto V and Leanza L: Targeting the mitochondrial potassium channel Kv1.3 to kill cancer cells: Drugs, strategies, and new perspectives. SLAS Discov. 24:882–892. 2019. View Article : Google Scholar : PubMed/NCBI

79 

Na W, Ma B, Shi S, Chen Y, Zhang H, Zhan Y and An H: Procyanidin B1, a novel and specific inhibitor of Kv10.1 channel, suppresses the evolution of hepatoma. Biochem Pharmacol. 178:1140892020. View Article : Google Scholar : PubMed/NCBI

80 

Zhao W, Bai B, Hong Z, Zhang X and Zhou B: Berbamine (BBM), a natural STAT3 inhibitor, synergistically enhances the antigrowth and proapoptotic effects of sorafenib on hepatocellular carcinoma cells. ACS Omega. 5:24838–24847. 2020. View Article : Google Scholar : PubMed/NCBI

81 

Zhou Q, Kwan HY, Chan HC, Jiang JL, Tam SC and Yao X: Blockage of voltage-gated K+ channels inhibits adhesion and proliferation of hepatocarcinoma cells. Int J Mol Med. 11:261–266. 2003.PubMed/NCBI

82 

Rosa P, Catacuzzeno L, Sforna L, Mangino G, Carlomagno S, Mincione G, Petrozza V, Ragona G, Franciolini F and Calogero A: BK channels blockage inhibits hypoxia-induced migration and chemoresistance to cisplatin in human glioblastoma cells. J Cell Physiol. 233:6866–6877. 2018. View Article : Google Scholar : PubMed/NCBI

83 

Wang X, Chen Y, Zhang Y, Guo S, Mo L, An H and Zhan Y: Eag1 voltage-dependent potassium channels: Structure, electrophysiological characteristics, and function in cancer. J Membr Biol. 250:123–132. 2017. View Article : Google Scholar : PubMed/NCBI

84 

García-Quiroz J and Camacho J: Astemizole: An old anti-histamine as a new promising anti-cancer drug. Anticancer Agents Med Chem. 11:307–314. 2011. View Article : Google Scholar : PubMed/NCBI

85 

de Guadalupe Chávez-López M, Pérez-Carreón JI, Zuñiga-García V, Díaz-Chávez J, Herrera LA, Caro-Sánchez CH, Acuña-Macías I, Gariglio P, Hernández-Gallegos E, Chiliquinga AJ and Camacho J: Astemizole-based anticancer therapy for hepatocellular carcinoma (HCC), and Eag1 channels as potential early-stage markers of HCC. Tumour Biol. 36:6149–6158. 2015. View Article : Google Scholar : PubMed/NCBI

86 

Roy AM, Baliga MS, Elmets CA and Katiyar SK: Grape seed proanthocyanidins induce apoptosis through p53, Bax, and caspase 3 pathways. Neoplasia. 7:24–36. 2005. View Article : Google Scholar : PubMed/NCBI

87 

Mantena SK and Katiyar SK: Grape seed proanthocyanidins inhibit UV-radiation-induced oxidative stress and activation of MAPK and NF-kappaB signaling in human epidermal keratinocytes. Free Radic Biol Med. 40:1603–1614. 2006. View Article : Google Scholar : PubMed/NCBI

88 

Mohr CJ, Steudel FA, Gross D, Ruth P, Lo WY, Hoppe R, Schroth W, Brauch H, Huber SM and Lukowski R: Cancer-associated intermediate conductance Ca2+-Activated K+ Channel KCa3.1. Cancers (Basel). 11:1092019. View Article : Google Scholar : PubMed/NCBI

89 

Catacuzzeno L, Fioretti B and Franciolini F: Expression and role of the intermediate-conductance calcium-activated potassium channel KCa3.1 in glioblastoma. J Signal Transduct. 2012:4215642012. View Article : Google Scholar : PubMed/NCBI

90 

Bulk E, Ay AS, Hammadi M, Ouadid-Ahidouch H, Schelhaas S, Hascher A, Rohde C, Thoennissen NH, Wiewrodt R, Schmidt E, et al: Epigenetic dysregulation of KCa 3.1 channels induces poor prognosis in lung cancer. Int J Cancer. 137:1306–1317. 2015. View Article : Google Scholar : PubMed/NCBI

91 

Toyama K, Wulff H, Chandy KG, Azam P, Raman G, Saito T, Fujiwara Y, Mattson DL, Das S, Melvin JE, et al: The intermediate-conductance calcium-activated potassium channel KCa3.1 contributes to atherogenesis in mice and humans. J Clin Invest. 118:3025–3037. 2008. View Article : Google Scholar : PubMed/NCBI

92 

Freise C, Ruehl M, Seehofer D, Hoyer J and Somasundaram R: The inhibitor of Ca(2+)-dependent K+ channels TRAM-34 blocks growth of hepatocellular carcinoma cells via downregulation of estrogen receptor alpha mRNA and nuclear factor-kappaB. Invest New Drugs. 31:452–457. 2013. View Article : Google Scholar : PubMed/NCBI

93 

Liu Y, Zhao L, Ma W, Cao X, Chen H, Feng D, Liang J, Yin K and Jiang X: The blockage of KCa3.1 channel inhibited proliferation, migration and promoted apoptosis of human hepatocellular carcinoma cells. J Cancer. 6:643–651. 2015. View Article : Google Scholar : PubMed/NCBI

94 

Freise C, Heldwein S, Erben U, Hoyer J, Köhler R, Jöhrens K, Patsenker E, Ruehl M, Seehofer D, Stickel F and Somasundaram R: K+-channel inhibition reduces portal perfusion pressure in fibrotic rats and fibrosis associated characteristics of hepatic stellate cells. Liver Int. 35:1244–1252. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Yang S, Yang S, Zhang H, Hua H, Kong Q, Wang J and Jiang Y: Targeting Na+/K+-ATPase by berbamine and ouabain synergizes with sorafenib to inhibit hepatocellular carcinoma. Br J Pharmacol. 178:4389–4407. 2021. View Article : Google Scholar : PubMed/NCBI

96 

Alevizopoulos K, Calogeropoulou T, Lang F and Stournaras C: Na+/K+ ATPase inhibitors in cancer. Curr Drug Targets. 15:988–1000. 2014. View Article : Google Scholar : PubMed/NCBI

97 

Simpson CD, Mawji IA, Anyiwe K, Williams MA, Wang X, Venugopal AL, Gronda M, Hurren R, Cheng S, Serra S, et al: Inhibition of the sodium potassium adenosine triphosphatase pump sensitizes cancer cells to anoikis and prevents distant tumor formation. Cancer Res. 69:2739–2747. 2009. View Article : Google Scholar : PubMed/NCBI

98 

Durlacher CT, Chow K, Chen XW, He ZX, Zhang X, Yang T and Zhou SF: Targeting Na+/K+-translocating adenosine triphosphatase in cancer treatment. Clin Exp Pharmacol Physiol. 42:427–443. 2015. View Article : Google Scholar : PubMed/NCBI

99 

Jiang W, Li G, Li W, Wang P, Xiu P, Jiang X, Liu B, Sun X and Jiang H: Sodium orthovanadate overcomes sorafenib resistance of hepatocellular carcinoma cells by inhibiting Na+/K+-ATPase activity and hypoxia-inducible pathways. Sci Rep. 8:97062018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2023
Volume 50 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen X, Zhang L, He L, Zheng L and Tuo B: Potassium channels as novel molecular targets in hepatocellular carcinoma (Review). Oncol Rep 50: 185, 2023
APA
Chen, X., Zhang, L., He, L., Zheng, L., & Tuo, B. (2023). Potassium channels as novel molecular targets in hepatocellular carcinoma (Review). Oncology Reports, 50, 185. https://doi.org/10.3892/or.2023.8622
MLA
Chen, X., Zhang, L., He, L., Zheng, L., Tuo, B."Potassium channels as novel molecular targets in hepatocellular carcinoma (Review)". Oncology Reports 50.4 (2023): 185.
Chicago
Chen, X., Zhang, L., He, L., Zheng, L., Tuo, B."Potassium channels as novel molecular targets in hepatocellular carcinoma (Review)". Oncology Reports 50, no. 4 (2023): 185. https://doi.org/10.3892/or.2023.8622