Pathophysiology of hepatic Na+/H+ exchange (Review)

  • Authors:
    • Tingting Li
    • Biguang Tuo
  • View Affiliations

  • Published online on: June 12, 2020     https://doi.org/10.3892/etm.2020.8888
  • Pages: 1220-1229
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Na+/H+ exchangers (NHEs) are a family of membrane proteins that contribute to exchanging one intracellular proton for one extracellular sodium. The family of NHEs consists of nine known members, NHE1‑9. Each isoform represents a different gene product that has unique tissue expression, membrane localization, physiological effects, pathological regulation and sensitivity to drug inhibitors. NHE1 was the first to be discovered and is often referred to as the ‘housekeeping’ isoform of the NHE family. NHEs are not only involved in a variety of physiological processes, including the control of transepithelial Na+ absorption, intracellular pH, cell volume, cell proliferation, migration and apoptosis, but also modulate complex pathological events. Currently, the vast majority of review articles have focused on the role of members of the NHE family in inflammatory bowel disease, intestinal infectious diarrhea and digestive system tumorigenesis, but only a few reviews have discussed the role of NHEs in liver disease. Therefore, the present review described the basic biology of NHEs and highlighted their physiological and pathological effects in the liver.

1. Introduction

Intracellular pH (pHi) is usually maintained between 6.9-7.2; it is important to maintain a normal pHi as a decrease in pHi may influence the normal functioning of proteins, ion channels and several other physiological processes involved in cell proliferation, division and differentiation (1). Some ion exchanger families regulate proton flux at the plasma membrane, such as Na+/H+ exchanger (NHE), Cl-/HCO3- exchanger, Na+/HCO3- cotransporter and Na+-driven Cl-/HCO3- exchanger (2). The present review focused on the NHE family. NHEs are pH-regulated membrane proteins that exchange extracellular Na+ for intracellular H+ with a stoichiometry of one for one (2). The inward Na+ gradient produced by Na+/K+-ATPase provides a constant driving force for H+ efflux (3). Moreover, NHEs are activated by decreases in pHi, and thus are likely to respond to an increase in proton load during acute acid stimulation (4).

NHEs are evolutionarily conserved membrane transporters; the solute carrier 9A (SLC9A) family contains the well-characterized plasma membrane and intracellular NHE isoforms (NHE1-9), and the SLC9B subgroup consists of Na+/H+ antiporter 1 and Na+/H+ antiporter 2(5). Different NHE isoforms are positioned differently. NHE1 is the ‘housekeeping’ isoform in NHE family (6) and is nearly ubiquitous in the plasma membrane of almost all tissues (7), while the other isoforms have more restricted localization and function. The specific localization of NHE1 may vary depending on cell type. In fibroblasts, NHE1 is mainly localized in lamellae and participates in migration and anchoring (8). However, in epithelial cells, NHE1 is distributed in the basolateral membrane (9). NHE2-5 are also localized to the plasma membrane, but have more restricted tissue distributions (10). For example, NHE2 is an apical membrane protein found mainly in the stomach and intestines (11). Both NHE3 and NHE4 are highly expressed in the kidney and gastrointestinal tract (12). In the gastrointestinal tract, NHE3 is mainly expressed in the intestine, while NHE4 is expressed in the stomach (12). Moreover, NHE5 is expressed primarily in the brain (13). The isoforms NHE6-NHE9 exist in intracellular organelles, where they participate in the maintenance of pHi (14). NHE6 is expressed in early recycling endosomes and mitochondria, NHE7 is located in the trans-Golgi network, NHE8 is in the mid- to trans-Golgi and NHE9 is expressed in late recovered endosomes (15,16).

Various subtypes of NHEs are related to the pathogenesis of digestive diseases, such as Barrett's esophagus (17), gastric cancer (18), IBD (19), colon cancer (20) or liver diseases. Currently, the vast majority of review articles have focused on the role of the NHE family members in IBD (21), intestinal infectious diarrhea (22) and digestive system tumorigenesis (23), and to the best of our knowledge, only a few have reported the role of NHEs in liver disease. The liver is the largest digestive gland in the human digestive system, and plays an important role in metabolism, deoxygenation, glycogen storage and secretory protein synthesis (24). Disease development in the liver seriously affects the normal function of the body (25). Thus, it is important to study the physiological and pathological regulation of the liver. However, the role of NHEs in liver function is not fully understood, although all NHEs except NHE5 have been detected in this organ (26).

Therefore, the present review details the physiology and pathology of NHEs in the liver, including the regulation of hepatocyte volume, hepatocyte growth, regeneration, proliferation, apoptosis, bile formation and other physiological activities. The pathologies discussed include non-alcoholic fatty liver disease (NAFLD), liver fibrosis, liver cancer and other liver diseases.

2. Structure and function of NHEs

The sequences of the nine subtypes of NHEs in the SLC9A subfamily are significantly different, with amino acid identities ranging between <12% (NHE1 vs. NHE9) and >70% (NHE6 vs. NHE7) (27). Despite these differences, silico-predicted transmembrane protein domains have suggested very similar structural arrangements for all nine isoforms, which have a high degree of similarity in the NH2-terminal hydrophobic domain, which contains multiple predicted membrane-spanning segments (27). However, it is important to note that NHE membrane topology has been most extensively studied in the NHE1 isoform (28). The complete membrane protein consists of 815 amino acids, and the first 500 amino acids of the protein are speculated to consist of 12 transmembrane hydrophobic domains (29). A C-terminal hydrophilic cytosolic domain of ~315 amino acids regulates the protein and mediates cytoskeletal interactions (30,31). Moreover, NHE2, 3, 4 and 5 have been reported to have 42, 39, 42 and 39% amino acid homology to NHE1, respectively (12). The NHE1 protein contains N- and O-glycosylated residues (32) and the N- and C- termini of NHE1 are found in the cytosol (6). Growth factors, hormones, integrins, osmotic stress and other signaling pathways regulate the activity of NHE1 via the mediation of the C-terminal domain, thus determining the pHi (33). In addition, there are some binding sites in the C-terminus, such as calmodulin (CaM), CaM homologous protein (CHP) and esrin/radixin/moesin (ERM) (4). When CaM binds to NHE1, it eliminates self-inhibition and activates NHE1. CHP AND ERM are bound to the cytosolic regulatory tail and also support the physiological activity of NHE1(4) (Fig. 1).

3. NHEs in hepatic physiological regulation

NHEs are the most widely studied pHi regulators in various animal cells, including hepatocytes (34). Intracellular acid load produced by normal hepatocyte metabolism activates NHE proteins to catalyze the electroneutral exchange of one extracellular Na+ with one intracellular H+, thus constituting a key component that prevents cell acidosis (4). Furthermore, this exchange process depends on the inward-directed Na+ gradient produced by the Na+/K+-ATPase to excrete H+ from the cytoplasm (9,35). NHEs are also involved in regulating the volume of hepatocytes (36), hepatocyte growth, regeneration, proliferation, apoptosis and bile formation, and a series of physiological activities, which are described below.

Regulation of cell volume is critical for liver function in healthy and disease states (37). Shrinkage or swelling of cells may result in disruption of the integrity of the cell membrane and cytoskeletal structure. To survive, ions must pass via certain ion transporters to avoid excessive changes in cell volume (38,39). When cells are exposed to hypertonic extracellular media, cell contraction triggers a regulated cell volume increase, which is largely accomplished by cellular ion uptake (40,41). Cellular contraction also stimulates Na+/K+/2Cl- cotransporters (NKCC) and/or NHEs in parallel with Cl-/HCO3- exchangers (40). H+ excretion via NHEs and HCO3- exiting via the Cl-/HCO3- exchanger are replenished in the cell by H2CO3, which is readily produced by CO2; this process achieves NaCl entry (42). On the other hand, Na+, which enters the cell via the NKCC and NHE, is pumped out by the Na+/K+-ATPase in exchange for K+, which eventually causes KCl uptake by the cells (42) (Fig. 2). Moreover, the NKCC isoforms NKCC1 and NKCC2(43) and the NHE isomers NHE1, 2 and 4 are activated by cell contraction, while NHE3 is inhibited by cell contraction (43).

The volume sensitivity of metabolism is an integral part of hormone signaling (44,45). The insulin involves changes in the volume of hepatocytes (42). In addition to activating NHEs and NKCC to expand hepatocyte volume and serving an antiproteolytic role, (42) insulin activates NHEs by binding to receptors linked to tyrosine kinases in hepatocytes and promotes the growth of hepatocytes (46). Similar to insulin, several growth factors, such as hepatocyte growth factor (HGF) (47,48), epidermal growth factor (EGF) (49) and transforming growth factor (TGF)-α (50), increase cell volume by stimulating NHEs, which is essential for stimulating cell proliferation (40). HGF is one of the most effective mitogens in hepatocytes and is often used to study the mechanism of hepatocyte proliferation (51). HGF, similar to insulin, activates NHEs in hepatocytes via a tyrosine kinase CaM-dependent pathway (51). EGF rapidly stimulates NHEs before increasing DNA synthesis, which not only promotes the proliferation of hepatocytes, but also participates in the regulation of liver regeneration (52). Dallenbach et al (53) measured pHi and homeostasis NHEs mRNA expression to compare the activity and regulation of NHEs in hepatocytes isolated after two-thirds partial hepatectomy or sham operation. These authors reported that during liver regeneration induced by partial hepatectomy in rats, NHEs in hepatocytes were activated at early, transient and posttranscriptional levels (53). The effect of liver regeneration on NHEs in hepatocytes is similar in qualitative and quantitative terms to that of hepatocytes exposed to EGF (54), suggesting that NHEs may participate in the regulation of liver regeneration as EGF in liver injury.

The effects of cell proliferation may be mediated by enhancing cell survival or inhibiting apoptosis (55). Intracellular acidification and cell volume reduction are markers of apoptosis, while NHE1 intracellular alkalization and the regulatory volume increases may be antiapoptotic signals (2,56). TGF-β, which induces apoptosis of hepatic parenchymal cells (57), has a significant inhibitory effect on NHE activity in short-term cultured rat hepatocytes, especially in cells isolated from perivenular regions, in which apoptosis is more frequently observed (58).

Hepatocytes and bile duct epithelial cells are involved in bile secretion and absorption (59). Bile duct epithelial cells also serve a role in the transport of water, electrolytes, sugar, bile and amino acids, and express several transport proteins to modify the primary production of hepatocyte bile (60). In biliary cells, four isoforms (NHE1-4) have been identified (61,62). Basolateral NHE1 is generally speculated to be involved in pHi, cell volume homeostasis and fluid and electrolyte transport, particularly secretin-induced bile secretion (63,64). In addition, NHE3 has been detected in cholangiocarcinoma cells in rats (65,66) and in gallbladder epithelial cells in calves (67) and prairie dogs (68). Targeted destruction of the NHE3 gene results in inhibition of fluid reabsorption in isolated bile duct units. For example, a study in mice found that decreased gallbladder absorption of bile may be the result of a decrease in NHE3 activity caused by an increased level of NHE3 phosphorylated at serine-552; this increase in phosphorylation is hypothesized to lead to a higher turnover of NHE3, which leads to a decrease in the gallbladder's concentrating function (69). Moreover, prairie dogs represent a good animal model for human gallstone formation, and their gallbladder epithelial cells exhibit H+ gradient-dependent Na+ uptake via NHE1 (~6% of total intake), NHE2 (~66% of total intake) and NHE3 (~28% of total intake), indicating a significant contribution of NHEs to epithelial Na+ absorption (68,70). Along with the findings showing increased absorption of Na+ and liquid in the early stage of gallstones (71), it has been proposed that apical membrane NHEs may be involved in the pathogenesis of gallstones (69). In conclusion, the aforementioned results suggested that decreases in NHE activity affects the absorption capacity of bile duct cells.

NHE1 may also regulate cell differentiation, as the absence or inhibition of NHE1 impairs the differentiation pathway (72). Furthermore, NHE1 function is important in cytoskeletal tissue and cell migration (22). The cytoplasmic tail of NHE1 acts as an anchor for actin filaments via the binding of ezrin, Radixin and moesin proteins, and the destruction of these interactions or inhibition of NHE1 activity leads to the inhibition of cell migration and the formation of external adhesions (2).

4. NHEs in hepatic pathology

Role of NHEs in NAFLD

NAFLD is closely related to liposome imbalance, and hepatic steatosis is considered to be the first stage in the development of NAFLD (73). With the development of fibrosis and inflammation, NAFLD can progress to non-alcoholic steatohepatitis (NASH) and eventually lead to liver fibrosis, cirrhosis and cancer (74,75). NHE activity is associated with steatosis in NAFLD. Previous studies (76) have compared the expression of NHE1 in the livers of normal diet mice and high-fat diet mice, and revealed that the expression of NHE1 in the livers of high-fat diet mice was nearly tripled and the long-term ablation of NHE1 activity in mice weakened high lipid diet-induced liver lipid accumulation, which suggests that NHE activity plays a role in the development of NAFLD (76). Farnesoid X receptor (FXR) is a nuclear hormone receptor. It has been reported that activation of FXR attenuates the development of hepatic steatosis (77,78), and FXR agonists reduce NASH-associated fibrosis (79,80). The prolipogenic liver X receptor (LXRa) is an important regulator of lipid metabolism. LXR activation promotes hepatic steatosis (81,82), and treatment with liver-specific LXR inhibitors reduced the development of hepatic steatosis (83). Prasad et al (76) revealed that in the livers of NHE1-null (KO) mice, increased expression of FXR and downregulation of LXRa expression were consistent with the results of long-term NHE1 deletion in reducing liver lipid accumulation induced by high-fat diet. In addition, the key regulators of adipogenesis, acetyl-CoA carboxylase α (Acc1) and Acc β (Acc2), are downregulated in NHE1-KO liver (76). Moreover, downregulation of Acc1 and Acc2 expression levels can reverse hepatic steatosis (84). Based on these findings, it was speculated that the loss of NHEs in the liver may lead to increased expression of FXR, downregulation of LXRa and downregulation of Acc1/Acc2 expression, which may reduce or reverse liver steatosis during the occurrence of fatty liver diseases such as NAFLD, which slows the development of NAFLD.

NHEs in liver fibrosis

Hepatic fibrosis is a common response to chronic liver injury of variable origins, such as viruses and metabolism (85). The mechanisms of liver fibrosis include activation of hepatic stellate cells (HSCs) and extracellular matrix (ECM) protein deposition, including various collagens and matrix glycoconjugates (86). During the development of fibrosis, HSCs proliferate and the activation process is characterized by the appearance of myofibroblast-like phenotypes that accumulate near necrotic areas (87). Activated HSCs are characterized by the expression of α-smooth muscle actin (SMA) (87), increased cell numbers, loss of retinoic acid (88,89) and increased expression of collagen fibrin (90). Currently, oxidative stress (91,92), paracrine stimulation of damaged hepatocytes, cytokines (93,94) and mitogens, such as platelet-derived growth factor (PDGF), TGF-β and insulin-like growth factor I, have been reported to promote the proliferation of HSCs and matrix synthesis. All of these factors activate NHEs, mostly likely the subtype 1, in the liver, and NHE activation is one of the earliest responses to mitogens and growth factors in most cell types (95). Previous studies have shown that NHE1 is the main pH regulator in HSCs, and its activity increases with the activation process of HSCs (96,97). When different growth factors and oxidative stress stimulate HSC proliferation and collagen type synthesis, the activity of NHE1 protein increases (96,98-100). Furthermore, the mechanism of HSCs proliferation is related to the increase in cell volume caused by NHE activation. For example, an increase in cell volume itself induces multiple changes in cellular function and gene expression by activating the osmosignaling pathway, and is a prerequisite for cell division and proliferation (38,101). It has also been reported that an increase in cell volume is parallel to the process via which fibroblasts transition from G1 to S phase (102).

Inhibition of NHEs selectively blocks ribonucleotide reductase, an enzyme that is critical for DNA synthesis (103)

The antioxidant resveratrol also inhibits nucleoside reductase activity (104), thus suggesting the possible role of reactive oxygen species as a common mechanism of NHE activation in HSCs; the promotion of fibrosis caused by oxidative stress is also partly due to NHE activation in HSCs (99). Benedetti et al (105) used the NHE inhibitors amiloride and cariporide, and revealed that these inhibited the proliferation and activation of NHE and HSCs by blocking PDGF and oxidative stress in vitro, suggesting that NHE inhibitors may reduce DNA synthesis in HSCs by inhibiting the activity of NHEs. Benedetti et al (105) also found that in vivo administration of amiloride and cariporide reduced the formation of α-SMA positive chains and the expression of type I procollagen in the local proliferation induced by two different liver injury models, dimethyl nitrosamine and bile duct ligation. Therefore, NHE appears to play a catalytic role in the pathogenesis of liver fibrosis (105). Collectively, it was speculated that the activity of NHEs may be closely related to the activation of HSC during liver fibrosis.

NHEs in liver cancer

Hepatocellular carcinoma (HCC) is one of the most common malignant tumors in the world and has a poor prognosis (106). The occurrence and development of cancer are closely related to dysregulation of cell energy metabolism, which is known as the Warburg effect (107). As a supportive anticancer therapy, glucose restriction (GR) inhibits enhanced glycolytic activity in cancer cells via energy-dependent signaling pathways, including the insulin like growth factor-1/PI3K/Akt/mTOR pathway (108). Previous studies have reported that intracellular alkalization is a major transformation event for cancer cells. For instance, one study showed that glioblastoma (MG) is a highly glycolytic malignant tumor that has a strong dependence on pH, and NHE1 activation drives cytoplasmic alkalization (109). However, inhibition of NHE1 in MG can acidify tumor cells, while healthy astrocytes are not affected; this finding may facilitate the development of treatment for malignant tumors (109). Targeting proton dynamics associated with pHi gradients has been proposed as a potential cancer prevention strategy and treatment (110). Kim et al demonstrated that curcumin treatment or GR slightly inhibited NHE1, while the combined treatment of curcumin and GR further enhanced the inhibitory effect on NHE1 and reduced pHi (110). Since the activation of NHE1 depends on energy and Akt (111), GR enhances the ability of curcumin to synergistically inhibit NHE1. Therefore, the combined treatment of GR and curcumin may have an important role in the regulation of pH in human hepatoma cells.

The expression of NHE1 in tumor tissues is not only related to the tumor size in HCC, but also venous invasion and pathological TNM staging (112). Previous studies have reported that inhibition of NHE1 blocks the invasion and metastasis of SMMC-7721 and HepG2 liver cancer cells (113,114). Invasion and migration of malignant tumor cells require destruction of the basement membrane and ECM proteolysis (115). The acidic extracellular pH (pHe) of tumor cells is also crucial in the activation of extracellular proteinases and the degradation of ECM in tumors (116,117). In the early stages of cell migration, invasion and metastasis, ECM decomposition is mainly mediated by matrix metalloproteinases (MMP)-2, -3 and -9 (114,118). Moreover, alkaline pHi promotes the expression of vascular endothelial growth factor (VEGF), which plays an important role in inducing neovascularization and promoting tumor growth and metastatic potential (Fig. 3) (119-121). Hypoxia is also a common feature of the tumor microenvironment and has been shown to stimulate invasion and metastasis (122,123). Hypoxia activates ERK1/2, a family of mitogen-activated protein kinases that play a major role in signaling pathways that are involved in cell scatter, motion, invasion, proliferation and survival (124). ERK1/2 also regulates the expression of MMPs and VEGF (125,126). Yang et al (114) demonstrated that inhibition of NHE1 by ethyl-isopropyl-amiloride (EIPA) inhibited HepG2 cell invasion and metastasis, and that EIPA inhibition acts by downregulating MMP-2, MMP-9 and VEGF in an ERK1/2-dependent manner. Moreover, NHE1 not only affects the migration and invasion of tumor cells, but is also related to the apoptosis of tumor cells (127). Bcl-2 family members, such as Bcl-2, Bcl-xL and Bax, play a crucial role in controlling apoptosis (127). Previous studies have reported that the NHE1 inhibitor EIPA downregulates Bcl-2, and upregulates Bax expression in HepG2 cells, leading to tumor cell apoptosis (128) (Fig. 3).

Interleukin 6 (IL6) is a key cytokine involved in the development and progression of inflammation-associated HCC. For example. Xu et al (128) found that IL6 activates the functional activity of NHE1, induces the interaction of NHE1, Na+/Ca2+ exchanger1 (NCX1) and calmodulin (CaM), and upregulates the expression of NHE1 in human hepatoma cells. Benzo[a]pyrene (B[a]P) is a prototype of polycyclic aromatic hydrocarbons, and is a human carcinogen (107). In addition to triggering apoptotic signals, B[a]P may induce survival signals and participate in the promotion of cancer (107). Previous studies have also reported that B[a]P induces metabolic reprogramming, which involves the activation of NHE1, and it leads to epithelial-mesenchymal transition (129-131). Ginsenoside Rg3, the main pharmacologically active compound extracted from Chinese ginseng, has been widely recognized as having antitumor properties in various cancer types (132,133), including inhibition of HCC cell proliferation, induction of apoptosis and inhibition of angiogenesis (134) and metastasis (135). Recently, it has been reported that Rg3 inhibits HCC cell proliferation and induces apoptosis by decreasing NHE1 expression and activity, and Rg3-mediated NHE1 inhibition is dependent on the EGF/EGFR/ERK1/2/hypoxia-inducible factor-1α signaling pathway (136). As a common ion transporter, NHEs are regulated by numerous substances. For example, IL6 and B[a]P can activate the activity of NHE1, while Rg3 can inhibit the activity of NHEs (107,128,132). Therefore, based on the aforementioned description of IL6, B[a]P and Rg3 in HCC, it was suggested that NHEs may be a potential therapeutic target in HCC. However, further studies are required to identify the potential mechanisms.

Cholangiocellular carcinoma (CCC) is the second most common primary liver cancer after HCC, with an increased incidence (137). Furthermore, chronic inflammation and oxidative stress play a key role in the development of CCC (138). NHEs form a potential link between controlling pHi and tumor development. Therefore, Elsing et al (62) determined the effect of oxidative stress on NHEs using tert-Butyl hydroperoxide (t-BOOH), a hydrogen peroxide, in the biliary epithelial cancer cell line Mz-Cha-1. These authors demonstrated that t-BOOH reduced NHE activity in a dose-dependent manner; at 4 mM t-BOOH, the NHE activity was almost absent and glutathione supplementation and intracellular Ca2+ chelation partially restored NHE activity. Moreover, in Mz-Cha-1 cells, inhibition of NHE by oxidative stress depends in part on the presence of intracellular Ca2+ and intracellular glutathione levels (62).

5. NHEs in other liver diseases

Hepatic failure (HF) is a life-threatening disease with a very high mortality rate (139), and hepatocyte apoptosis leading to HF is an important event in hepatocyte death. Tumor necrosis factor-α (TNF-α) is an inflammatory factor and an inducer of hepatocyte apoptosis (140). It has been reported that TNF-α induces NHE activity in hepatocytes in a time-dependent manner (141). Activation of NHEs increases intracellular Na+, promotes Na+/Ca2+ exchange and causes Ca2+ overload (142), which is considered to be the key factor in cell damage. Moreover, an increase in intracellular Ca2+ concentration automatically activates calpain, a calcium-dependent protease (143). The antiapoptotic family member Bcl-xL is a natural substrate for calpain, and NHE mediates TNF-α-induced hepatocyte apoptosis via Ca2+/calpain-dependent degradation of Bcl-xL (141). However, the NHE inhibitor cariporide reverses the effects induced by TNF-α and has a protective effect on acute HF (141).

Endotoxin-mediated production of proinflammatory cytokines plays an important role in the pathogenesis of liver disease (144). Previous studies have reported that lipopolysaccharide (LPS) causes liver damage by increasing the release of TNF-α in a NASH model (145). Therefore, interfering with LPS-induced inflammatory responses may help to alleviate inflammation associated with liver disease. Heat shock proteins (Hsp70) play an important role in LPS-mediated inflammatory responses (144). NHE1 is considered to be a mediator of inflammatory responses in macrophages (146-148) and has been reported to interact with Hsp70(149). Inhibition of Hsp70 substrate binding activity in vivo reduces induction of proinflammatory factors (144). Huang et al (144) treated macrophages and livers with LPS and revealed a significant increase in the association of NHE1-Hsp70, suggesting that the formation of the NHE1-Hsp70 complex is essential for the induction of proinflammatory factors. Therefore, LPS-induced liver damage may be prevented by disrupting the NHE1-Hsp70 interaction.

In addition, the beneficial effects of the NHE inhibitor EIPA in blocking NHEs in a partial hepatic ischemia rat model suggested a positive role for NHE1 in oxidative liver injury, and indicated that inhibition of NHEs is a potential strategy for preventing or reducing ischemic liver injury (27).

6. Conclusion

NHEs are ion transporters that are widely present in a variety of organisms and are important regulators at the cellular, tissue and systemic levels (150). To the best of our knowledge, the current review is the first detailed description of the physiology and pathology of NHEs in the liver. While NHEs are common targets for various inflammatory stimuli, the effect of selective targeted therapy of NHEs in the liver are inconclusive, and thus further studies are required. Numerous experimental models currently show that NHE inhibitors lack major toxic effects, and several, such as cariporide, have been used in preclinical and clinical trials (151). However, a large number of studies have only analyzed the effects of single factors, and have not considered that their function in physiological and pathological conditions may mainly be the result of the interaction of various transporters. Therefore, more comprehensive methods are required to alter the function, and regulate and target NHEs in liver-related pathology.

Acknowledgements

The authors would like to thank Professor Jiaxing An (Department of Gastroenterology, Affiliated Hospital to Zunyi Medical University) for their highly professional services.

Funding

This study was supported by grant from the Clinical Medical Research Center for Digestive Diseases,Guizhou province,China

Availability of data and materials

Not applicable.

Authors' contributions

TL wrote the manuscript and participated in information collection, analysis, organization and figure design. BT primarily revised and final editing. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Kraut JA and Madias NE: Treatment of acute metabolic acidosis: A pathophysiologic approach. Nat Rev Nephrol. 8:589–601. 2012.PubMed/NCBI View Article : Google Scholar

2 

Putney LK, Denker SP and Barber DL: The changing face of the Na+/H+ exchanger, NHE1: Structure, regulation, and cellular actions. Annu Rev Pharmacol Toxicol. 42:527–552. 2002.PubMed/NCBI View Article : Google Scholar

3 

Kondapalli KC, Prasad H and Rao R: An inside job: How endosomal Na(+)/H(+) exchangers link to autism and neurological disease. Front Cell Neurosci. 8(172)2014.PubMed/NCBI View Article : Google Scholar

4 

Malo ME and Fliegel L: Physiological role and regulation of the Na+/H+ exchanger. Can J Physiol Pharmacol. 84:1081–1095. 2006.PubMed/NCBI View Article : Google Scholar

5 

Gurney MA, Laubitz D, Ghishan FK and Kiela PR: Pathophysiology of Intestinal Na+/H+ exchange. Cell Mol Gastroenterol Hepatol. 3:27–40. 2017.PubMed/NCBI View Article : Google Scholar

6 

Sardet C, Franchi A and Pouysségur J: Molecular cloning, primary structure, and expression of the human growth factor-activatable Na+/H+ antiporter. Cell. 56:271–280. 1989.PubMed/NCBI View Article : Google Scholar

7 

Xu H, Chen H, Li J, Zhao Y and Ghishan FK: Disruption of NHE8 expression impairs Leydig cell function in the testes. Am J Physiol Cell Physiol. 308:C330–C338. 2015.PubMed/NCBI View Article : Google Scholar

8 

Denker SP and Barber DL: Cell migration requires both ion translocation and cytoskeletal anchoring by the Na-H exchanger NHE1. J Cell Biol. 159:1087–1096. 2002.PubMed/NCBI View Article : Google Scholar

9 

Orlowski J and Grinstein S: Diversity of the mammalian sodium/proton exchanger SLC9 gene family. Pflugers Arch. 447:549–565. 2004.PubMed/NCBI View Article : Google Scholar

10 

Zhao H, Carney KE, Falgoust L, Pan JW, Sun D and Zhang Z: Emerging roles of Na+/H+ exchangers in epilepsy and developmental brain disorders. Prog Neurobiol. 138-140:19–35. 2016.PubMed/NCBI View Article : Google Scholar

11 

Malakooti J, Dahdal RY, Schmidt L, Layden TJ, Dudeja PK and Ramaswamy K: Molecular cloning, tissue distribution, and functional expression of the human Na(+)/H(+) exchanger NHE2. Am J Physiol. 277:G383–G390. 1999.PubMed/NCBI View Article : Google Scholar

12 

Loo SY, Chang MK, Chua CS, Kumar AP, Pervaiz S and Clement MV: NHE-1: A promising target for novel anti-cancer therapeutics. Curr Pharm Des. 18:1372–1382. 2012.PubMed/NCBI View Article : Google Scholar

13 

Kurata T, Rajendran V, Fan S, Ohta T, Numata M and Fushida S: NHE5 regulates growth factor signaling, integrin trafficking, and degradation in glioma cells. Clin Exp Metastasis. 36:527–538. 2019.PubMed/NCBI View Article : Google Scholar

14 

Pescosolido MF, Stein DM, Schmidt M, El Achkar CM, Sabbagh M, Rogg JM, Tantravahi U, McLean RL, Liu JS, Poduri A, et al: Genetic and phenotypic diversity of NHE6 mutations in Christianson syndrome. Ann Neurol. 76:581–593. 2014.PubMed/NCBI View Article : Google Scholar

15 

Nakamura N, Tanaka S, Teko Y, Mitsui K and Kanazawa H: Four Na+/H+ exchanger isoforms are distributed to Golgi and post-Golgi compartments and are involved in organelle pH regulation. J Biol Chem. 280:1561–1572. 2005.PubMed/NCBI View Article : Google Scholar

16 

Ko M, Quiñones-Hinojosa A and Rao R: Emerging links between endosomal pH and cancer. Cancer Metastasis Rev: Apr 6, 2020 (Epub ahead of print).

17 

Laczkó D, Rosztóczy A, Birkás K, Katona M, Rakonczay Z Jr, Tiszlavicz L, Róka R, Wittmann T, Hegyi P and Venglovecz V: Role of ion transporters in the bile acid-induced esophageal injury. Am J Physiol Gastrointest Liver Physiol. 311:G16–G31. 2016.PubMed/NCBI View Article : Google Scholar

18 

Hosogi S, Miyazaki H, Nakajima K, Ashihara E, Niisato N, Kusuzaki K and Marunaka Y: An inhibitor of Na(+)/H(+) exchanger (NHE), ethyl isopropyl amiloride (EIPA), diminishes proliferation of MKN28 human gastric cancer cells by decreasing the cytosolic Cl(-) concentration via DIDS sensitive pathways. Cell Physiol Biochem. 30:1241–1253. 2012.PubMed/NCBI View Article : Google Scholar

19 

Khan I and Khan K: Uncoupling of Carbonic Anhydrase from Na H exchanger 1 in Experimental Colitis: A Possible Mechanistic Link with Na H Exchanger. Biomolecules. 9(700)2019.PubMed/NCBI View Article : Google Scholar

20 

Xu H, Li J, Chen H and Ghishan FK: NHE8 Deficiency Promotes Colitis-Associated Cancer in Mice via Expansion of Lgr5-Expressing Cells. Cell Mol Gastroenterol Hepatol. 7:19–31. 2018.PubMed/NCBI View Article : Google Scholar

21 

Magalhães D, Cabral JM, Soares-da-Silva P and Magro F: Role of epithelial ion transports in inflammatory bowel disease. Am J Physiol Gastrointest Liver Physiol. 310:G460–G476. 2016.PubMed/NCBI View Article : Google Scholar

22 

Das S, Jayaratne R and Barrett KE: The role of ion transporters in the pathophysiology of infectious diarrhea. Cell Mol Gastroenterol Hepatol. 6:33–45. 2018.PubMed/NCBI View Article : Google Scholar

23 

Cao L, Yuan Z, Liu M and Stock C: (Patho-)Physiology of Na+/H+ Exchangers (NHEs) in the Digestive System. Front Physiol. 10(1566)2020.PubMed/NCBI View Article : Google Scholar

24 

Yong W: Image diagnosis of common liver lesions Image research and medical application(on the column). J Imaging Res Med Applic, 2018 (In Chinese).

25 

Lowry SF and Brennan MF: Abnormal liver function during parenteral nutrition: Relation to infusion excess. J Surg Res. 26:300–307. 1979.

26 

Laohapitakworn S, Thongbunchoo J, Nakkrasae LI, Krishnamra N and Charoenphandhu N: Parathyroid hormone (PTH) rapidly enhances CFTR-mediated HCO3- secretion in intestinal epithelium-like Caco-2 monolayer: A novel ion regulatory action of PTH. Am J Physiol Cell Physiol. 301:C137–C149. 2011.PubMed/NCBI View Article : Google Scholar

27 

Xu H, Ghishan FK and Kiela PR: SLC9 Gene Family: Function, expression, and regulation. Compr Physiol. 8:555–583. 2018.PubMed/NCBI View Article : Google Scholar

28 

Kemp G, Young H and Fliegel L: Structure and function of the human Na+/H+ exchanger isoform 1. Channels (Austin). 2:329–336. 2008.PubMed/NCBI View Article : Google Scholar

29 

Fuster DG and Alexander RT: Traditional and emerging roles for the SLC9 Na+/H+ exchangers. Pflugers Arch. 466:61–76. 2014.PubMed/NCBI View Article : Google Scholar

30 

Stock C and Schwab A: Role of the Na/H exchanger NHE1 in cell migration. Acta Physiol (Oxf). 187:149–157. 2006.PubMed/NCBI View Article : Google Scholar

31 

Fliegel L: Structural and Functional Changes in the Na(+)/H(+) Exchanger Isoform 1, Induced by Erk1/2 Phosphorylation. Int J Mol Sci. 20(2378)2019.PubMed/NCBI View Article : Google Scholar

32 

Fuster DG and Alexander RT: Traditional and emerging roles for the SLC9 Na+/H+ exchangers. Pflugers Arch. 466:61–76. 2014.PubMed/NCBI View Article : Google Scholar

33 

Amith SR and Fliegel L: Regulation of the Na+/H+ Exchanger (NHE1) in Breast Cancer Metastasis. Cancer Res. 73:1259–1264. 2013.PubMed/NCBI View Article : Google Scholar

34 

Lee CH, Cragoe EJ Jr and Edwards AM: Control of hepatocyte DNA synthesis by intracellular pH and its role in the action of tumor promoters. J Cell Physiol. 195:61–69. 2003.PubMed/NCBI View Article : Google Scholar

35 

Aharonovitz O, Zaun HC, Balla T, York JD, Orlowski J and Grinstein S: Intracellular pH regulation by Na(+)/H(+) exchange requires phosphatidylinositol 4,5-bisphosphate. J Cell Biol. 150:213–224. 2000.PubMed/NCBI View Article : Google Scholar

36 

Ahmed KH, Pelster B and Krumschnabel G: Signalling pathways involved in hypertonicity- and acidification-induced activation of Na+/H+ exchange in trout hepatocytes. J Exp Biol. 209:3101–3113. 2006.PubMed/NCBI View Article : Google Scholar

37 

Haussinger D: Osmosensing and osmosignaling in the liver. Wiener medizinische Wochenschrift (1946). 158:549–552. 2008.PubMed/NCBI View Article : Google Scholar

38 

Lang F, Busch GL, Ritter M, Völkl H, Waldegger S, Gulbins E and Häussinger D: Functional significance of cell volume regulatory mechanisms. Physiol Rev. 78:247–306. 1998.PubMed/NCBI View Article : Google Scholar

39 

Jo AO, Ryskamp DA, Phuong TT, Verkman AS, Yarishkin O, MacAulay N and Križaj D: TRPV4 and AQP4 channels synergistically regulate cell volume and calcium homeostasis in retinal Müller Glia. J Neurosci. 35:13525–13537. 2015.PubMed/NCBI View Article : Google Scholar

40 

Lang F, Föller M, Lang K, Lang P, Ritter M, Vereninov A, Szabo I, Huber SM and Gulbins E: Cell volume regulatory ion channels in cell proliferation and cell death. Methods Enzymol. 428:209–225. 2007.PubMed/NCBI View Article : Google Scholar

41 

Mongin AA: Volume-regulated anion channel - a frenemy within the brain. Pflugers Arch. 468:421–441. 2016.PubMed/NCBI View Article : Google Scholar

42 

Lang PA, Graf D, Boini KM, Lang KS, Klingel K, Kandolf R and Lang F: Cell volume, the serum and glucocorticoid inducible kinase 1 and the liver. Z Gastroenterol. 49:713–719. 2011.PubMed/NCBI View Article : Google Scholar

43 

Hoffmann EK, Lambert IH and Pedersen SF: Physiology of cell volume regulation in vertebrates. Physiol Rev. 89:193–277. 2009.PubMed/NCBI View Article : Google Scholar

44 

Häussinger D and Lang F: Cell volume and hormone action. Trends Pharmacol Sci. 13:371–373. 1992.PubMed/NCBI View Article : Google Scholar

45 

Lee MJ: Hormonal regulation of adipogenesis. Compr Physiol. 7:1151–1195. 2017.PubMed/NCBI View Article : Google Scholar

46 

O'Connor McCourt M, Soley M, Hayden LJ and Hollenberg MD: Receptors for epidermal growth factor (urogastrone) and insulin in primary cultures of rat hepatocytes maintained in serum free medium. Biochem Cell Biol. 64:803–810. 1986.PubMed/NCBI View Article : Google Scholar

47 

Dykes SS, Steffan JJ and Cardelli JA: Lysosome trafficking is necessary for EGF-driven invasion and is regulated by p38 MAPK and Na+/H+ exchangers. BMC Cancer. 17(672)2017.PubMed/NCBI View Article : Google Scholar

48 

Steffan JJ, Williams BC, Welbourne T and Cardelli JA: HGF-induced invasion by prostate tumor cells requires anterograde lysosome trafficking and activity of Na+-H+ exchangers. J Cell Sci. 123:1151–1159. 2010.PubMed/NCBI View Article : Google Scholar

49 

Coaxum SD, Blanton MG, Joyner A, Akter T, Bell PD, Luttrell LM, Raymond JR Sr, Lee MH, Blichmann PA, Garnovskaya MN, et al: Epidermal growth factor-induced proliferation of collecting duct cells from Oak Ridge polycystic kidney mice involves activation of Na+/H+ exchanger. Am J Physiol Cell Physiol. 307:C554–C560. 2014.PubMed/NCBI View Article : Google Scholar

50 

Mead JE and Fausto N: Transforming growth factor alpha may be a physiological regulator of liver regeneration by means of an autocrine mechanism. Proc Natl Acad Sci USA. 86:1558–1562. 1989.PubMed/NCBI View Article : Google Scholar

51 

Kaneko A, Hayashi N, Tanaka Y, Horimoto M, Ito T, Sasaki Y, Fusamoto H and Kamada T: Activation of Na+/H+ exchanger by hepatocyte growth factor in hepatocytes. Hepatology. 22:629–636. 1995.PubMed/NCBI

52 

Goodrich AL and Suchy FJ: Na(+)-H+ exchange in basolateral plasma membrane vesicles from neonatal rat liver. Am J Physiol. 259:G334–G339. 1990.PubMed/NCBI View Article : Google Scholar

53 

Dällenbach A, Marti U and Renner EL: Hepatocellular Na+/H+ exchange is activated early, transiently and at a posttranscriptional level during rat liver regeneration. Hepatology. 19:1290–1301. 1994.PubMed/NCBI View Article : Google Scholar

54 

Moule SK and McGivan JD: Epidermal growth factor and cyclic AMP stimulate Na+/H+ exchange in isolated rat hepatocytes. Eur J Biochem. 187:677–682. 1990.PubMed/NCBI View Article : Google Scholar

55 

Love MR, Palee S, Chattipakorn SC and Chattipakorn N: Effects of electrical stimulation on cell proliferation and apoptosis. J Cell Physiol. 233:1860–1876. 2018.PubMed/NCBI View Article : Google Scholar

56 

Cardoso VG, Gonçalves GL, Costa-Pessoa JM, Thieme K, Lins BB, Casare FAM, de Ponte MC, Camara NOS and Oliveira-Souza M: Angiotensin II-induced podocyte apoptosis is mediated by endoplasmic reticulum stress/PKC-δ/p38 MAPK pathway activation and trough increased Na+/H+ exchanger isoform 1 activity. BMC Nephrol. 19(179)2018.PubMed/NCBI View Article : Google Scholar

57 

Gaitantzi H, Meyer C, Rakoczy P, Thomas M, Wahl K, Wandrer F, Bantel H, Alborzinia H, Wölfl S, Ehnert S, et al: Ethanol sensitizes hepatocytes for TGF-β-triggered apoptosis. Cell Death Dis. 9(51)2018.PubMed/NCBI View Article : Google Scholar

58 

Benedetti A, Di Sario A, Svegliati Baroni G and Jezequel AM: Transforming growth factor beta 1 increases the number of apoptotic bodies and decreases intracellular pH in isolated periportal and perivenular rat hepatocytes. Hepatology. 22:1488–1498. 1995.PubMed/NCBI

59 

Martínez-Ansó E, Castillo JE, Díez J, Medina JF and Prieto J: Immunohistochemical detection of chloride/bicarbonate anion exchangers in human liver. Hepatology. 19:1400–1406. 1994.PubMed/NCBI

60 

Marin JJ, Macias RI, Briz O, Banales JM and Monte MJ: Bile Acids in Physiology, Pathology and Pharmacology. Curr Drug Metab. 17:4–29. 2015.PubMed/NCBI View Article : Google Scholar

61 

Marti U, Elsing C, Renner EL, Liechti-Gallati S and Reichen J: Differential expression of Na+ H(+)-antiporter mRNA in biliary epithelial cells and in hepatocytes. J Hepatol. 24:498–502. 1996.PubMed/NCBI View Article : Google Scholar

62 

Elsing C, Voss A, Herrmann T, Kaiser I, Huebner CA and Schlenker T: Oxidative stress reduces Na+/H+ exchange (NHE) activity in a biliary epithelial cancer cell line (Mz-Cha-1). Anticancer Res. 31:459–465. 2011.PubMed/NCBI

63 

Hirata K and Nathanson MH: Bile duct epithelia regulate biliary bicarbonate excretion in normal rat liver. Gastroenterology. 121:396–406. 2001.PubMed/NCBI View Article : Google Scholar

64 

Hübner C, Stremmel W and Elsing C: Sodium, hydrogen exchange type 1 and bile ductular secretory activity in the guinea pig. Hepatology. 31:562–571. 2000.PubMed/NCBI View Article : Google Scholar

65 

Roussa E, Bertram J, Berge KE, Labori KJ, Thévenod F and Raeder MG: Differential regulation of vacuolar H+ -ATPase and Na+/H+ exchanger 3 in rat cholangiocytes after bile duct ligation. Histochem Cell Biol. 125:419–428. 2006.PubMed/NCBI View Article : Google Scholar

66 

Mennone A, Biemesderfer D, Negoianu D, Yang CL, Abbiati T, Schultheis PJ, Shull GE, Aronson PS and Boyer JL: Role of sodium/hydrogen exchanger isoform NHE3 in fluid secretion and absorption in mouse and rat cholangiocytes. Am J Physiol Gastrointest Liver Physiol. 280:G247–G254. 2001.PubMed/NCBI View Article : Google Scholar

67 

Bazzini C, Bottà G, Meyer G, Baroni MD and Paulmichl M: The presence of NHE1 and NHE3 Na+-H+ exchangers and an apical cAMP-independent Cl- channel indicate that both absorptive and secretory functions are present in calf gall bladder epithelium. Exp Physiol. 86:571–583. 2001.PubMed/NCBI View Article : Google Scholar

68 

Narins SC, Park EH, Ramakrishnan R, Garcia FU, Diven JN, Balin BJ, Hammond CJ, Sodam BR, Smith PR and Abedin MZ: Functional characterization of Na(+)/H(+) exchangers in primary cultures of prairie dog gallbladder. J Membr Biol. 197:123–134. 2004.PubMed/NCBI View Article : Google Scholar

69 

Chen Y, Wu S, Tian Y and Kong J: Phosphorylation and subcellular localization of Na+/H+ exchanger isoform 3 (NHE3) are associated with altered gallbladder absorptive function after formation of cholesterol gallstones. J Physiol Biochem. 73:133–139. 2017.

70 

Saier MH Jr, Yen MR, Noto K, Tamang DG and Elkan C: The Transporter Classification Database: Recent advances. Nucleic Acids Res. 37 (Database):D274–D278. 2009.PubMed/NCBI View Article : Google Scholar

71 

Giurgiu DI, Saunders-Kirkwood KD, Roslyn JJ and Abedin MZ: Sequential changes in biliary lipids and gallbladder ion transport during gallstone formation. Ann Surg. 225:382–390. 1997.PubMed/NCBI View Article : Google Scholar

72 

Li X, Karki P, Lei L, Wang H and Fliegel L: Na+/H+ exchanger isoform 1 facilitates cardiomyocyte embryonic stem cell differentiation. Am J Physiol Heart Circ Physiol. 296:H159–H170. 2009.PubMed/NCBI View Article : Google Scholar

73 

Manne V, Handa P and Kowdley KV: Pathophysiology of Nonalcoholic Fatty Liver Disease/Nonalcoholic Steatohepatitis. Clin Liver Dis. 22:23–37. 2018.PubMed/NCBI View Article : Google Scholar

74 

Baffy G, Brunt EM and Caldwell SH: Hepatocellular carcinoma in non-alcoholic fatty liver disease: An emerging menace. J Hepatol. 56:1384–1391. 2012.PubMed/NCBI View Article : Google Scholar

75 

Friedman SL, Neuschwander-Tetri BA, Rinella M and Sanyal AJ: Mechanisms of NAFLD development and therapeutic strategies. Nat Med. 24:908–922. 2018.PubMed/NCBI View Article : Google Scholar

76 

Prasad V, Chirra S, Kohli R and Shull GE: NHE1 deficiency in liver: Implications for non-alcoholic fatty liver disease. Biochem Biophys Res Commun. 450:1027–1031. 2014.PubMed/NCBI View Article : Google Scholar

77 

Cipriani S, Mencarelli A, Palladino G and Fiorucci S: FXR activation reverses insulin resistance and lipid abnormalities and protects against liver steatosis in Zucker (fa/fa) obese rats. J Lipid Res. 51:771–784. 2010.PubMed/NCBI View Article : Google Scholar

78 

Ma Y, Huang Y, Yan L, Gao M and Liu D: Synthetic FXR agonist GW4064 prevents diet-induced hepatic steatosis and insulin resistance. Pharm Res. 30:1447–1457. 2013.PubMed/NCBI View Article : Google Scholar

79 

Mudaliar S, Henry RR, Sanyal AJ, Morrow L, Marschall HU, Kipnes M, Adorini L, Sciacca CI, Clopton P, Castelloe E, et al: Efficacy and safety of the farnesoid X receptor agonist obeticholic acid in patients with type 2 diabetes and nonalcoholic fatty liver disease. Gastroenterology. 145:574–82.e1. 2013.PubMed/NCBI View Article : Google Scholar

80 

Tully DC, Rucker PV, Chianelli D, Williams J, Vidal A, Alper PB, Mutnick D, Bursulaya B, Schmeits J, Wu X, et al: Discovery of tropifexor (LJN452), a highly potent non-bile acid FXR agonist for the treatment of cholestatic liver diseases and nonalcoholic steatohepatitis (NASH). J Med Chem. 60:9960–9973. 2017.PubMed/NCBI View Article : Google Scholar

81 

Baranowski M, Zabielski P, Blachnio Zabielska AU, Harasim E, Chabowski A and Gorski J: Insulin sensitizing effect of LXR agonist T0901317 in high fat fed rats is associated with restored muscle GLUT4 expression and insulin stimulated AS160 phosphorylation. Cell Physiol Biochem. 33:1047–1057. 2014.PubMed/NCBI View Article : Google Scholar

82 

Ducheix S, Montagner A, Theodorou V, Ferrier L and Guillou H: The liver X receptor: A master regulator of the gut-liver axis and a target for non alcoholic fatty liver disease. Biochem Pharmacol. 86:96–105. 2013.PubMed/NCBI View Article : Google Scholar

83 

Griffett K, Solt LA, El-Gendy BD, Kamenecka TM and Burris TP: A liver-selective LXR inverse agonist that suppresses hepatic steatosis. ACS Chem Biol. 8:559–567. 2013.PubMed/NCBI View Article : Google Scholar

84 

Savage DB, Choi CS, Samuel VT, Liu ZX, Zhang D, Wang A, Zhang XM, Cline GW, Yu XX, Geisler JG, et al: Reversal of diet-induced hepatic steatosis and hepatic insulin resistance by antisense oligonucleotide inhibitors of acetyl-CoA carboxylases 1 and 2. J Clin Invest. 116:817–824. 2006.PubMed/NCBI View Article : Google Scholar

85 

Dewidar B, Meyer C, Dooley S and Meindl Beinker AN: TGF beta in hepatic stellate cell activation and liver fibrogenesis-Updated 2019. Cells. 8(1419)2019.PubMed/NCBI View Article : Google Scholar

86 

Yang C, Zeisberg M, Mosterman B, Sudhakar A, Yerramalla U, Holthaus K, Xu L, Eng F, Afdhal N and Kalluri R: Liver fibrosis: Insights into migration of hepatic stellate cells in response to extracellular matrix and growth factors. Gastroenterology. 124:147–159. 2003.PubMed/NCBI View Article : Google Scholar

87 

Li J, Zhao YR and Tian Z: Roles of hepatic stellate cells in acute liver failure: From the perspective of inflammation and fibrosis. World J Hepatol. 11:412–420. 2019.PubMed/NCBI View Article : Google Scholar

88 

Tsuchida T and Friedman SL: Mechanisms of hepatic stellate cell activation. Nat Rev Gastroenterol Hepatol. 14:397–411. 2017.PubMed/NCBI View Article : Google Scholar

89 

Mak KM, Leo MA and Lieber CS: Alcoholic liver injury in baboons: Transformation of lipocytes to transitional cells. Gastroenterology. 87:188–200. 1984.PubMed/NCBI

90 

Chen Z, Jain A, Liu H, Zhao Z and Cheng K: Targeted drug delivery to hepatic stellate cells for the treatment of liver fibrosis. J Pharmacol Exp Ther. 370:695–702. 2019.PubMed/NCBI View Article : Google Scholar

91 

Lan T, Kisseleva T and Brenner DA: Deficiency of NOX1 or NOX4 prevents liver inflammation and fibrosis in mice through inhibition of hepatic stellate cell activation. PLoS One. 10(e0129743)2015.PubMed/NCBI View Article : Google Scholar

92 

Ou Q, Weng Y, Wang S, Zhao Y, Zhang F, Zhou J and Wu X: Silybin alleviates hepatic steatosis and fibrosis in NASH mice by inhibiting oxidative stress and involvement with the Nf-κB Pathway. Dig Dis Sci. 63:3398–3408. 2018.PubMed/NCBI View Article : Google Scholar

93 

Syn WK, Agboola KM, Swiderska M, Michelotti GA, Liaskou E, Pang H, Xie G, Philips G, Chan IS, Karaca GF, et al: NKT-associated hedgehog and osteopontin drive fibrogenesis in non-alcoholic fatty liver disease. Gut. 61:1323–1329. 2012.PubMed/NCBI View Article : Google Scholar

94 

Syn WK, Choi SS, Liaskou E, Karaca GF, Agboola KM, Oo YH, Mi Z, Pereira TA, Zdanowicz M, Malladi P, et al: Osteopontin is induced by hedgehog pathway activation and promotes fibrosis progression in nonalcoholic steatohepatitis. Hepatology. 53:106–115. 2011.PubMed/NCBI View Article : Google Scholar

95 

Grinstein S, Rotin D and Mason MJ: Na+/H+ exchange and growth factor-induced cytosolic pH changes. Role in cellular proliferation. Biochim Biophys Acta. 988:73–97. 1989.PubMed/NCBI View Article : Google Scholar

96 

Di Sario A, Baroni GS, Bendia E, D'Ambrosio L, Ridolfi F, Marileo JR, Jezequel AM and Benedetti A: Characterization of ion transport mechanisms regulating intracellular pH in hepatic stellate cells. Am J Physiol. 273:G39–G48. 1997.PubMed/NCBI View Article : Google Scholar

97 

Trappoliere M, Caligiuri A, Schmid M, Bertolani C, Failli P, Vizzutti F, Novo E, di Manzano C, Marra F, Loguercio C, et al: Silybin, a component of sylimarin, exerts anti-inflammatory and anti-fibrogenic effects on human hepatic stellate cells. J Hepatol. 50:1102–1111. 2009.PubMed/NCBI View Article : Google Scholar

98 

Di Sario A, Bendia E, Svegliati Baroni G, Ridolfi F, Bolognini L, Feliciangeli G, Jezequel AM, Orlandi F and Benedetti A: Intracellular pathways mediating Na+/H+ exchange activation by platelet-derived growth factor in rat hepatic stellate cells. Gastroenterology. 116:1155–1166. 1999.PubMed/NCBI View Article : Google Scholar

99 

Svegliati-Baroni G, Di Sario A, Casini A, Ferretti G, D'Ambrosio L, Ridolfi F, Bolognini L, Salzano R, Orlandi F and Benedetti A: The Na+/H+ exchanger modulates the fibrogenic effect of oxidative stress in rat hepatic stellate cells. J Hepatol. 30:868–875. 1999.PubMed/NCBI View Article : Google Scholar

100 

Svegliati Baroni G, D'Ambrosio L, Ferretti G, Casini A, Di Sario A, Salzano R, Ridolfi F, Saccomanno S, Jezequel AM and Benedetti A: Fibrogenic effect of oxidative stress on rat hepatic stellate cells. Hepatology. 27:720–726. 1998.PubMed/NCBI View Article : Google Scholar

101 

Häussinger D and Schliess F: Osmotic induction of signaling cascades: Role in regulation of cell function. Biochem Biophys Res Commun. 255:551–555. 1999.PubMed/NCBI View Article : Google Scholar

102 

Pendergrass WR, Angello JC, Kirschner MD and Norwood TH: The relationship between the rate of entry into S phase, concentration of DNA polymerase alpha, and cell volume in human diploid fibroblast-like monokaryon cells. Exp Cell Res. 192:418–425. 1991.PubMed/NCBI View Article : Google Scholar

103 

Vairo G, Cocks BG, Cragoe EJ Jr and Hamilton JA: Selective suppression of growth factor-induced cell cycle gene expression by Na+/H+ antiport inhibitors. J Biol Chem. 267:19043–19046. 1992.PubMed/NCBI

104 

Fontecave M, Lepoivre M, Elleingand E, Gerez C and Guittet O: Resveratrol, a remarkable inhibitor of ribonucleotide reductase. FEBS Lett. 421:277–279. 1998.PubMed/NCBI View Article : Google Scholar

105 

Benedetti A, Di Sario A, Casini A, Ridolfi F, Bendia E, Pigini P, Tonnini C, D'Ambrosio L, Feliciangeli G, Macarri G, et al: Inhibition of the NA(+)/H(+) exchanger reduces rat hepatic stellate cell activity and liver fibrosis: An in vitro and in vivo study. Gastroenterology. 120:545–556. 2001.PubMed/NCBI View Article : Google Scholar

106 

Huang Q, Li J, Zheng J and Wei A: The carcinogenic role of the Notch signaling pathway in the development of hepatocellular carcinoma. J Cancer. 10:1570–1579. 2019.PubMed/NCBI View Article : Google Scholar

107 

Hardonnière K, Saunier E, Lemarié A, Fernier M, Gallais I, Héliès-Toussaint C, Mograbi B, Antonio S, Bénit P, Rustin P, et al: The environmental carcinogen benzo[a]pyrene induces a Warburg-like metabolic reprogramming dependent on NHE1 and associated with cell survival. Sci Rep. 6(30776)2016.PubMed/NCBI View Article : Google Scholar

108 

Thyfault JP and Rector RS: Exercise Combats Hepatic Steatosis: Potential Mechanisms and Clinical Implications. Diabetes. 69:517–524. 2020.PubMed/NCBI View Article : Google Scholar

109 

Harguindey S, Polo Orozco J, Alfarouk KO and Devesa J: Hydrogen ion dynamics of cancer and a new molecular, biochemical and metabolic approach to the etiopathogenesis and treatment of brain malignancies. Int J Mol Sci. 20(4278)2019.PubMed/NCBI View Article : Google Scholar

110 

Kim SW, Cha MJ, Lee SK, Song BW, Jin X, Lee JM, Park JH and Lee JD: Curcumin treatment in combination with glucose restriction inhibits intracellular alkalinization and tumor growth in hepatoma cells. Int J Mol Sci. 20(2375)2019.PubMed/NCBI View Article : Google Scholar

111 

Meima ME, Webb BA, Witkowska HE and Barber DL: The sodium-hydrogen exchanger NHE1 is an Akt substrate necessary for actin filament reorganization by growth factors. J Biol Chem. 284:26666–26675. 2009.PubMed/NCBI View Article : Google Scholar

112 

Yang X, Wang D, Dong W, Song Z and Dou K: Over-expression of Na+/H+ exchanger 1 and its clinicopathologic significance in hepatocellular carcinoma. Med Oncol. 27:1109–1113. 2010.PubMed/NCBI View Article : Google Scholar

113 

Yang X, Wang D, Dong W, Song Z and Dou K: Suppression of Na+/H+ exchanger 1 by RNA interference or amiloride inhibits human hepatoma cell line SMMC-7721 cell invasion. Med Oncol. 28:385–390. 2011.PubMed/NCBI View Article : Google Scholar

114 

Yang X, Wang D, Dong W, Song Z and Dou K: Inhibition of Na(+)/H(+) exchanger 1 by 5-(N-ethyl-N-isopropyl) amiloride reduces hypoxia-induced hepatocellular carcinoma invasion and motility. Cancer Lett. 295:198–204. 2010.PubMed/NCBI View Article : Google Scholar

115 

He X, Lee B and Jiang Y: Cell-ECM Interactions in Tumor Invasion. Adv Exp Med Biol. 936:73–91. 2016.PubMed/NCBI View Article : Google Scholar

116 

Stüwe L, Müller M, Fabian A, Waning J, Mally S, Noël J, Schwab A and Stock C: pH dependence of melanoma cell migration: Protons extruded by NHE1 dominate protons of the bulk solution. J Physiol. 585:351–360. 2007.PubMed/NCBI View Article : Google Scholar

117 

Reshkin SJ, Cardone RA and Harguindey S: : Na+-H+ exchanger, pH regulation and cancer. Recent Patents Anticancer Drug Discov. 8:85–99. 2013.PubMed/NCBI View Article : Google Scholar

118 

Keurhorst D, Liashkovich I, Frontzek F, Nitzlaff S, Hofschröer V, Dreier R and Stock C: MMP3 activity rather than cortical stiffness determines NHE1-dependent invasiveness of melanoma cells. Cancer Cell Int. 19(285)2019.PubMed/NCBI View Article : Google Scholar

119 

He B, Deng C, Zhang M, Zou D and Xu M: Reduction of intracellular pH inhibits the expression of VEGF in K562 cells after targeted inhibition of the Na+/H+ exchanger. Leuk Res. 31:507–514. 2007.PubMed/NCBI View Article : Google Scholar

120 

Apte RS, Chen DS and Ferrara N: VEGF in Signaling and Disease: Beyond Discovery and Development. Cell. 176:1248–1264. 2019.PubMed/NCBI View Article : Google Scholar

121 

Alfarouk KO: Tumor metabolism, cancer cell transporters, and microenvironmental resistance. J Enzyme Inhib Med Chem. 31:859–866. 2016.PubMed/NCBI View Article : Google Scholar

122 

Gilkes DM, Semenza GL and Wirtz D: Hypoxia and the extracellular matrix: Drivers of tumour metastasis. Nat Rev Cancer. 14:430–439. 2014.PubMed/NCBI View Article : Google Scholar

123 

Vaupel P, Kallinowski F and Okunieff P: Blood flow, oxygen and nutrient supply, and metabolic microenvironment of human tumors: A review. Cancer Res. 49:6449–6465. 1989.PubMed/NCBI

124 

Sun Y, Liu WZ, Liu T, Feng X, Yang N and Zhou HF: Signaling pathway of MAPK/ERK in cell proliferation, differentiation, migration, senescence and apoptosis. J Recept Signal Transduct Res. 35:600–604. 2015.PubMed/NCBI View Article : Google Scholar

125 

Chen LC, Shibu MA, Liu CJ, Han CK, Ju DT, Chen PY, Viswanadha VP, Lai CH, Kuo WW and Huang CY: ERK1/2 mediates the lipopolysaccharide-induced upregulation of FGF-2, uPA, MMP-2, MMP-9 and cellular migration in cardiac fibroblasts. Chem Biol Interact. 306:62–69. 2019.PubMed/NCBI View Article : Google Scholar

126 

Wang JC, Chen SY, Wang M, Ko JL, Wu CL, Chen CC, Lin HW and Chang YY: Nickel-induced VEGF expression via regulation of Akt, ERK1/2, NFκB, and AMPK pathways in H460 cells. Environ Toxicol. 34:652–658. 2019.PubMed/NCBI View Article : Google Scholar

127 

Yang X, Wang D, Dong W, Song Z and Dou K: Expression and modulation of Na(+) /H(+) exchanger 1 gene in hepatocellular carcinoma: A potential therapeutic target. J Gastroenterol Hepatol. 26:364–370. 2011.PubMed/NCBI View Article : Google Scholar

128 

Xu J, Ji B, Wen G, Yang Y, Jin H, Liu X, Xie R, Song W, Song P, Dong H, et al: Na+/H+ exchanger 1, Na+/Ca2+ exchanger 1 and calmodulin complex regulates interleukin 6-mediated cellular behavior of human hepatocellular carcinoma. Carcinogenesis. 37:290–300. 2016.PubMed/NCBI View Article : Google Scholar

129 

Huc L, Sparfel L, Rissel M, Dimanche-Boitrel MT, Guillouzo A, Fardel O and Lagadic-Gossmann D: Identification of Na+/H+ exchange as a new target for toxic polycyclic aromatic hydrocarbons. FASEB J. 18:344–346. 2004.PubMed/NCBI View Article : Google Scholar

130 

Hardonnière K, Saunier E, Lemarié A, Fernier M, Gallais I, Héliès-Toussaint C, Mograbi B, Antonio S, Bénit P, Rustin P, et al: The environmental carcinogen benzo[a]pyrene induces a Warburg-like metabolic reprogramming dependent on NHE1 and associated with cell survival. Sci Rep. 6(30776)2016.PubMed/NCBI View Article : Google Scholar

131 

Dendelé B, Tekpli X, Hardonnière K, Holme JA, Debure L, Catheline D, Arlt VM, Nagy E, Phillips DH, Ovrebø S, et al: Protective action of n-3 fatty acids on benzo[a]pyrene-induced apoptosis through the plasma membrane remodeling-dependent NHE1 pathway. Chem Biol Interact. 207:41–51. 2014.PubMed/NCBI View Article : Google Scholar

132 

Wang J, Tian L, Khan MN, Zhang L, Chen Q, Zhao Y, Yan Q, Fu L and Liu J: Ginsenoside Rg3 sensitizes hypoxic lung cancer cells to cisplatin via blocking of NF-κB mediated epithelial-mesenchymal transition and stemness. Cancer Lett. 415:73–85. 2018.PubMed/NCBI View Article : Google Scholar

133 

Zhang C, Liu L, Yu Y, Chen B, Tang C and Li X: Antitumor effects of ginsenoside Rg3 on human hepatocellular carcinoma cells. Mol Med Rep. 5:1295–1298. 2012.PubMed/NCBI View Article : Google Scholar

134 

Zhou B, Wang J and Yan Z: Ginsenoside Rg3 attenuates hepatoma VEGF overexpression after hepatic artery embolization in an orthotopic transplantation hepatocellular carcinoma rat model. OncoTargets Ther. 7:1945–1954. 2014.PubMed/NCBI View Article : Google Scholar

135 

Zhou B, Yan Z, Liu R, Shi P, Qian S, Qu X, Zhu L, Zhang W and Wang J: Prospective Study of Transcatheter Arterial Chemoembolization (TACE) with Ginsenoside Rg3 versus TACE Alone for the Treatment of Patients with Advanced Hepatocellular Carcinoma. Radiology. 280:630–639. 2016.PubMed/NCBI View Article : Google Scholar

136 

Li X, Tsauo J, Geng C, Zhao H, Lei X and Li X: Ginsenoside Rg3 Decreases NHE1 Expression via Inhibiting EGF-EGFR-ERK1/2-HIF-1 [Formula: see text] Pathway in Hepatocellular Carcinoma: A Novel Antitumor Mechanism. Am J Chin Med. 46:1915–1931. 2018.PubMed/NCBI View Article : Google Scholar

137 

Kloeckner R, Ruckes C, Kronfeld K, Wörns MA, Weinmann A, Galle PR, Lang H, Otto G, Eichhorn W, Schreckenberger M, et al: Selective internal radiotherapy (SIRT) versus transarterial chemoembolization (TACE) for the treatment of intrahepatic cholangiocellular carcinoma (CCC): study protocol for a randomized controlled trial. Trials. 15(311)2014.PubMed/NCBI View Article : Google Scholar

138 

Uchida D, Takaki A, Ishikawa H, Tomono Y, Kato H, Tsutsumi K, Tamaki N, Maruyama T, Tomofuji T, Tsuzaki R, et al: Oxidative stress balance is dysregulated and represents an additional target for treating cholangiocarcinoma. Free Radic Res. 50:732–743. 2016.PubMed/NCBI View Article : Google Scholar

139 

Grek A and Arasi L: Acute liver failure. AACN Adv Crit Care. 27:420–429. 2016.PubMed/NCBI View Article : Google Scholar

140 

Ezquerro S, Mocha F, Frühbeck G, Guzmán-Ruiz R, Valentí V, Mugueta C, Becerril S, Catalán V, Gómez-Ambrosi J, Silva C, et al: Ghrelin Reduces TNF-α-Induced Human Hepatocyte Apoptosis, Autophagy, and Pyroptosis: Role in Obesity-Associated NAFLD. J Clin Endocrinol Metab. 104:21–37. 2019.PubMed/NCBI View Article : Google Scholar

141 

Liu Z, Wang S, Zhou H, Yang Y and Zhang M: Na+/H+ exchanger mediates TNF-alpha-induced hepatocyte apoptosis via the calpain-dependent degradation of Bcl-xL. J Gastroenterol Hepatol. 24:879–885. 2009.PubMed/NCBI View Article : Google Scholar

142 

Alexander RT, Dimke H and Cordat E: Proximal tubular NHEs: Sodium, protons and calcium? Am J Physiol Renal Physiol. 305:F229–F236. 2013.PubMed/NCBI View Article : Google Scholar

143 

Shi M, Zhang T, Sun L, Luo Y, Liu DH, Xie ST, Song XY, Wang GF, Chen XL, Zhou BC and Zhang YZ: Calpain, Atg5 and Bak play important roles in the crosstalk between apoptosis and autophagy induced by influx of extracellular calcium. Apoptosis. 18:435–451. 2013.PubMed/NCBI View Article : Google Scholar

144 

Huang C, Wang J, Chen Z, Wang Y and Zhang W: 2-phenylethynesulfonamide Prevents Induction of Pro-inflammatory Factors and Attenuates LPS-induced Liver Injury by Targeting NHE1-Hsp70 Complex in Mice. PLoS One. 8(e67582)2013.PubMed/NCBI View Article : Google Scholar

145 

Ceccarelli S, Panera N, Mina M, Gnani D, De Stefanis C, Crudele A, Rychlicki C, Petrini S, Bruscalupi G, Agostinelli L, et al: LPS-induced TNF-α factor mediates pro-inflammatory and pro-fibrogenic pattern in non-alcoholic fatty liver disease. Oncotarget. 6:41434–41452. 2015.PubMed/NCBI View Article : Google Scholar

146 

Liu CL, Zhang X, Liu J, Wang Y, Sukhova GK, Wojtkiewicz GR, Liu T, Tang R, Achilefu S, Nahrendorf M, et al: Na+-H+ exchanger 1 determines atherosclerotic lesion acidification and promotes atherogenesis. Nat Commun. 10(3978)2019.PubMed/NCBI View Article : Google Scholar

147 

Guan X, Hasan MN, Begum G, Kohanbash G, Carney KE, Pigott VM, Persson AI, Castro MG, Jia W and Sun D: Blockade of Na/H exchanger stimulates glioma tumor immunogenicity and enhances combinatorial TMZ and anti-PD-1 therapy. Cell Death Dis. 9(1010)2018.PubMed/NCBI View Article : Google Scholar

148 

Rotstein OD, Houston K and Grinstein S: Control of cytoplasmic pH by Na+/H+ exchange in rat peritoneal macrophages activated with phorbol ester. FEBS Lett. 215:223–227. 1987.PubMed/NCBI View Article : Google Scholar

149 

Ye Y, Jia X, Bajaj M and Birnbaum Y: Dapagliflozin Attenuates Na+/H+ Exchanger-1 in Cardiofibroblasts via AMPK Activation. Cardiovasc Drugs Ther. 32:553–558. 2018.PubMed/NCBI View Article : Google Scholar

150 

Ryuichi O, Masafumi M and Hiroshi K: Localization, ion transport activity, and physiological function of mammalian organellar NHEs. Seikagaku. J Jpn Biochem Soc. 82:2010.PubMed/NCBI(In Japanese).

151 

Karmazyn M: Pharmacology and clinical assessment of cariporide for the treatment coronary artery diseases. Expert Opin Investig Drugs. 9:1099–1108. 2000.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

August-2020
Volume 20 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li T and Li T: Pathophysiology of hepatic Na+/H+ exchange (Review). Exp Ther Med 20: 1220-1229, 2020
APA
Li, T., & Li, T. (2020). Pathophysiology of hepatic Na+/H+ exchange (Review). Experimental and Therapeutic Medicine, 20, 1220-1229. https://doi.org/10.3892/etm.2020.8888
MLA
Li, T., Tuo, B."Pathophysiology of hepatic Na+/H+ exchange (Review)". Experimental and Therapeutic Medicine 20.2 (2020): 1220-1229.
Chicago
Li, T., Tuo, B."Pathophysiology of hepatic Na+/H+ exchange (Review)". Experimental and Therapeutic Medicine 20, no. 2 (2020): 1220-1229. https://doi.org/10.3892/etm.2020.8888