
The role of Na,K‑ATPase in lung diseases (Review)
- Authors:
- Published online on: August 27, 2025 https://doi.org/10.3892/mmr.2025.13665
- Article Number: 300
-
Copyright: © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
The Na,K-ATPase was first described by Skou (1) in 1957. It is classified as a member of the P-type ATPase class, in this class, the enzyme is phosphorylated by ATP in the presence of Mg2+ and Na+ and dephosphorylated in the presence of K+ (2,3). As a heteromeric transmembrane protein, Na,K-ATPase consists of three subunits: The α subunit, β subunit and γ subunit (FXYD2). The α subunit, which serves as the catalytic unit, has four isoforms (α1, α2, α3 and α4). The β subunit, a highly glycosylated transmembrane protein, has three isoforms (β1, β2 and β3). As a regulatory subunit, it carries out multiple functions, including enhancing the translation efficiency and stability of the α subunit, assisting in the folding of the α subunit and facilitating the transport of the α subunit from the endoplasmic reticulum to the plasma membrane. Additionally, it regulates the affinity of Na+ and K+ for the enzyme and acts as a cell adhesion molecule (4–8). The γ subunit, belongs to the FXYD family, which has seven isoforms (FXYD1-7), is present in different tissues, such as kidney tissues, cardiac tissues and skeletal muscle, carries out a regulatory role by modulating the affinity of Na+ and K+ for the enzyme, influencing pump kinetics and transport properties and stabilizing Na,K-ATPase (9–12).
The Na,K-ATPase functions primarily as an ion pump, importing two K+ ions and exporting three Na+ ions at the expense of one ATP molecule in eukaryotic cells. This process contributes to maintaining high intracellular K+ levels, preserving the osmotic balance, providing energy for active transport and regulating the membrane potential (2,13–15). In addition, Na,K-ATPase acts as a ligand for cardiac glycosides and regulates multiple signaling pathways, including non-receptor tyrosine kinase gene (Src), PI3K, caveolin-1, protein phosphatase 2, EGFR and MAPK, thereby modulating cell proliferation, motility and apoptosis (Fig. 1) (16–20).
Numerous studies have indicated that Na,K-ATPase is involved in various diseases (21–23). The present review discusses the relationship between Na,K-ATPase and lung diseases.
Lung cancer
Lung cancer is one of the most frequently diagnosed types of cancer and a leading cause of cancer-related mortalities worldwide with ~2.2 million new cases and 1.76 million mortalities per year (24). Patients have varying 5-year survival rates from 4 to 17% depending on stage and regional differences (25). There are two main types of lung cancer: Non-small and small cell lung carcinomas (SLCs). The most common is non-small cell lung cancer (NSCLC), which represents >80% of cases. NSCLC is commonly subdivided into three subtypes: Squamous cell carcinoma, large cell carcinoma and adenocarcinoma. Adenocarcinoma has become the most common subtype of lung cancer globally in 2022, with sex differences diminishing (26). NSCLC is a heterogeneous cancer with cellular and genetic diversity. Clinical treatment has been based on molecular genotyping in NSCLC tumors, such as in epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase, ros proto-oncogene 1, receptor tyrosine kinase, rearranged during transfection and mesenchymal-epithelial transition factor (27).
Although new drugs for cancer are continuously being developed, the development of cancer therapies remains a considerable challenge due to chemoresistance. Na,K-ATPase has been studied as a target for cancer treatment, with cardiac steroids (CSs) binding to Na,K-ATPase and triggering several cell signaling pathways that result in anticancer effects by inhibiting proliferation and promoting autophagy or apoptosis (28–31). These effects vary depending on the cell type as well as the type and concentration of CS, and different cells frequently express different pump isoforms. At high concentrations, CSs were revealed to exhibit proapoptotic effects via Na,K-ATPase inhibition, which is not directly dependent on other intracellular cancer-specific pathways and kills cells by disrupting ionic homeostasis. However, at low concentrations, CSs are not selective for cancer cells and activate the Src-EGFR-Ras-Raf-kinase pathway through Na,K-ATPase, inhibiting the proliferation and survival of tumor cells (30,31). Src is a non-receptor tyrosine kinase that tightly associates with the Na,K-ATPase transmembrane complex in its activated form. Src activation contributes to MEK1/2 and ERK1/2 activation upon CG treatment and largely contributes to CG toxicity. Considering the well-described effects of CGs on endosomal trafficking, it is tempting to hypothesize that any activation of endosomal cycling by CGs modulates Src recruitment, signalosome assembly and subsequent cell survival mechanisms (32).
Smoking is a key risk factor for lung cancer and is usually associated with SCLC and squamous-cell carcinoma. Smoking impairs Na,K-ATPase activity through reactive oxygen species (ROS), although it does not notably alter protein expression of ROS. In patients with lung cancer who smoke, reduced expression levels of the Na,K-ATPase β1 subunit are associated with poor survival (33). Furthermore, studies have reported the downregulation of the Na,K-ATPase β1 subunit in other tumors, such as renal cell carcinoma and bladder cancer (34–36). The Na,K-ATPase β1 subunit serves as a tumor suppressor, maintaining epithelial polarity, enhancing cell-cell aggregation, reducing cell motility and invasiveness, and inhibiting cancer cell proliferation in vivo (19,37–42).
Overactivation of the α1-Na,K-ATPase/Src signalosome by oxidative stress during pathological states results in the phosphorylation of the Src kinase (pSrc), which activates the PI3K-Akt-mTOR pathway downstream. Activation of the PI3K-Akt pathway promotes the expression of the anti-apoptotic protein survivin, driving abnormal cell growth, survival, proliferation, angiogenesis and metabolism. Signalosome normalization using an inhibitory peptide resets apoptotic activity in malignant cells, with a significant decrease in tumor burden in vivo (43). Furthermore, a reduction in Na,K-ATPase α1 subunit expression can impair the proliferation and migration of A549 cells in vitro (44). Collectively, α1-NKA may serve as a novel target for clinical therapy. Additionally, FXYD3, antigen-8 of mammary tumor, is known to exert protection of the β1 subunit against glutathionylation, an oxidative modification that destabilizes the α-β heterodimer and inhibits Na,K-ATPase activity. The cellular expression of FXYD proteins that have the specific Cys residue is protective against oxidative stress-induced β1 subunit glutathionylation and Na+/K+-ATPase inhibition, while mutating the residue eliminates the protective effect of the expressed protein (45). FXYD3 proteins confer chemotherapy resistance when they are overexpressed in cancer cells. Chemotherapeutic compounds, such as doxorubicin, can induce oxidative stress and a study revealed that the suppression of FXYD3 with siRNA in pancreatic and breast cancer cells, which express FXYD3, increased doxorubicin-induced cytotoxicity (46). The point mutation in the cysteine residue of FXYD3 increases sensitivity to oxidative stress induced by the chemotherapeutic doxorubicin (47).
However, reduced expression of FXYD3 was reported in lung cancer cells, in which its inactivation was identified as a key mediator in lung cancer progression (48). In another study, the increasing levels of doublecortin-like kinase 1 in lung tumors, promotes the proliferation and metastasis of lung cancer cells through the downregulation of FXYD3 (49). FXYD3 regulates the activity of the Na,K-ATPase pump and may carry out a role in regulating cell proliferation, migration and invasion, key processes in cancer growth and metastasis (50).
The PI3K-Akt pathway has been reported to be constitutively activated in several types of malignancy (51). Factors that activate this pathway include loss of the tumor suppressor PTEN, activation/amplification of PI3K-Akt, activation of growth factor receptors including EGFR and exposure to carcinogenic agents (52). Digoxigenin, the aglycon of digoxin, binds to PI3K, could effectively treat breast cancer (53). Bufalin, an Na,K-ATPase ligand, exerts anti-cancer effects by inhibiting the proliferation of A549 cells and induces their apoptosis by suppressing the PI3K/Akt pathway and increasing BAX expression (54). Ouabain, another Na,K-ATPase ligand, promotes apoptosis in H292 cells by inducing the expression and activation of the apoptotic protein caspase 3 and degrading the anti-apoptotic protein Mcl-1 (55). Moreover, Na,K-ATPase α1/ouabain signaling facilitates the fine control of PD-L1 expression and dampens tumoral immunity (56). Perillyl alcohol, an Na,K-ATPase inhibitor, reduces the growth of NSCLC cells by inducing G2/M arrest, causing DNA damage, decreasing the mitochondrial transmembrane potential, increasing the accumulation of ROS and activating MAP kinases (57). Coimmunoprecipitation and immunolocalization studies have demonstrated the interaction between Na,K-ATPase and Bcl-XL and Bak in lung cancer cell lines, specifically fibrolamellar carcinoma and A549 cells (58,59). Na,K-ATPase, in its intrinsic role or as a receptor for CS, may serve as a potential target for lung cancer treatment.
COVID-19
Newly emerging viral diseases have become a major public health threat globally in recent years (60). During the last two decades, outbreaks of several viral diseases have been reported including the severe acute respiratory syndrome coronavirus (SARS-CoV) in 2002, H1N1 influenza in 2009, the Middle East respiratory syndrome coronavirus (MERS-CoV) in 2012, Ebola virus disease (EVD) in 2013 and Zika virus in 2015 (61). The outbreak of the novel coronavirus was initially reported to the World Health Organization (WHO) on December 31, 2019. On January 30, 2020, the WHO declared this viral outbreak as a public health emergency of international concern (62). As of December 21, 2022 >650 million individuals had coronavirus disease 2019 (COVID-19 and 6.6 million mortalities have been reported globally (63). COVID-19 is a potentially life-threatening condition caused by severe acute respiratory syndrome virus 2 (SARS-CoV-2), ~2% of infected patients and 15% of those who required hospitalization developed acute respiratory distress syndrome (ARDS), which is associated with poor outcomes (64,65).
Numerous SARS-CoV-2-mediated and COVID-19-related signals directly or indirectly impair the function of Na,K-ATPase, potentially contributing to alveolar epithelial barrier failure leading to further aggravation and persistence of lung injury (66,67). Deregulation of the Na,K-ATPase may also lead to extrapulmonary manifestations of COVID-19. The evidence suggests that various molecular mechanisms that are key in the regulation of transcription, translation, maturation and trafficking of the Na,K-ATPase are negatively regulated by SARS-CoV-2, leading to decreased expression of the a1, a2, a3, b1 and b3 subunits. Thus, Na,K-ATPase carries out an important role in delaying disease progression (68,69). Moreover, SARS-CoV-2 affects alveolar type II epithelial cells, which markedly disturb surfactant homeostasis and deprives the Na,K-ATPase of ATP, which gradually decreases alveolar gaseous exchange, resulting in intracellular hypoxic conditions and increased ROS. This hypoxia/ROS activates AMP-activated kinase, which further inhibits Na,K-ATPase and exacerbates lung deterioration (70,71). Oxidation-induced carbonylation of the Na,K-ATPase α1 subunit desensitizes Na,K-ATPase signaling, and the subsequent increased activity of Src results in increased ROS and inflammation, which can further promote the inflammatory cytokine storm (72). Mechanistically, SARS-CoV-2 spike proteins inhibited the PI3K/AKT/mTOR pathway by upregulating intracellular ROS levels, thus promoting the autophagic response. ROS production leads to the activation of signaling cascades, including JAK/STATs, PI3K/AKT and MAPK/ERK. Alternatively, elevated ROS levels can also inhibit the PI3K/AKT/mTOR pathway, thus enhancing autophagy levels (73). Upregulated expression of this pathway has been observed in several diseases of neoplastic, autoimmune and viral origin and is considered to be implicated in the pathogenesis of the disease. This has led to the hypothesis that single or dual inhibitors of this pathway could ameliorate the course of SARS-CoV-2 infection (74).
Hypoxia-inducible factor 1α (HIF1α) is an important activator of glycolysis and the inflammatory response and enhances virus infection and proinflammatory responses (75). Digoxin has been reported to suppress the expression of HIF1α, which may provide a more effective therapeutic target for both cancer and SARS-CoV-2 infection (76). BiP, also referred to as 78-Kilodalton glucose-regulated protein (GRP78), is upregulated both intracellularly and at the cell surface during SARS-CoV-2 infection. Oleandrin, a unique lipid-soluble CG derived from Nerium oleander, has a strong affinity for the α3 isoform of Na,K-ATPase. Oleandrin suppresses GRP78 induction and viral production without affecting cell viability and enhances the activity of antiviral agent therapy (77,78). It also produces an anti-inflammatory response through the activation of Nrf-2 antioxidant genes, which may be beneficial for preventing hyperinflammatory responses to infection with SARS-CoV-2 (79). Increasing evidence has revealed that dexamethasone is beneficial in patients with severe and critical COVID-19 and is well known to increase the activity of Na,K-ATPase, which may contribute to the beneficial effects of corticosteroid therapy (80,81). Thus, the upregulation of Na,K-ATPase may help control the progression of COVID-19, providing a new option for treatment.
ARDS
ARDS is a clinical syndrome of acute hypoxemic respiratory failure due to lung inflammation, not caused by cardiogenic pulmonary edema. It was first described in 1967 (82,83). Since then, the clinical definition of ARDS has been revised several times, first by an American-European consensus conference convened in 1992 by the American Thoracic Society and the European Society of Intensive Care Medicine (84) and subsequently by the ARDS Definition Task Force convened in Berlin in 2012 by the European Society of Intensive Care Medicine (85).
ARDS is characterized by an acute disruption of the alveolar-capillary barrier that leads to flooding of the alveolar space and thus, to marked impairment of gas exchange and ARDS is associated with a poor clinical outcome (86,87). Impairment of the enzyme Na,K-ATPase during ARDS not only prevents the resolution of lung oedema but also intensifies its formation (88). To clear pulmonary edema effectively, it is essential that a tight alveolar epithelial barrier is reestablished and that an Na+ gradient across the alveolar epithelium is created, which then drives the reabsorption of water from the alveolar space (89). Impaired alveolar fluid clearance (AFC) is one of the key mechanisms involved in this condition. AFC primarily depends on the basolaterally located Na,K-ATPase and the apically located epithelial sodium channel (ENaC) in epithelial alveolar cells. During the progression of ARDS, Na,K-ATPase is downregulated by various factors, including sepsis, hypoxia and influenza infection (90–92). Oleic acid (18:1 n-9), the most common and abundant fatty acid in the body of healthy individuals, is an Na,K-ATPase inhibitor and an Na+ channel inhibitor in the lung, resulting in notably increased endothelial permeability and inducing injury to lipid body formation in leukocytes and eicosanoid production (93,94). Consequently, Na,K-ATPase carries out a key role in the resolution of pulmonary edema (95).
Evidence demonstrated that the Src family kinase acted upstream of and was a required step (or trigger) for activation of both the PI3K/Akt and MAPK/ERK1/2 pathways, each of which is required for upregulation of alveolar epithelial cells, Na,K-ATPase activity and cell surface expression (96). MAPK is a key signal transduction system by which cells transduce extracellular signals into intracellular responses. Several reports suggest an important role of the MAPK signal transduction pathway in the regulation of Na,K-ATPase. β-Adrenergic agonists as well as dopamine stimulate MAPK activation that results in (long-term, >12 h) upregulation of Na,K-ATPase (97). Specialized proresolving mediators, as proinflammatory mediators, not only inhibit proinflammatory cytokine production and prevent leukocyte infiltration but also increase the ENaC, Na,K-ATPase and cystic fibrosis transmembrane conductance regulator (CFTR) levels to increase the AFC in patients with ARDS (98). Maresin1 is a newly described macrophage-derived mediator of inflammation resolution that stimulates AFC by upregulating Na,K-ATPase expression through the ALX/PI3K/Nedd4-2 pathway, suggesting that Maresin1 may constitute a novel therapeutic approach for the resolution of ARDS (99). Resolvin conjugates in tissue regeneration 1 (RCTR1) is an endogenous lipid mediator derived from docosahexaenoic acid that increases the protein levels of Na channels and Na,K-ATPase activity to improve AFC in acute lung injury (ALI) through the ALX/cAMP/PI3K/Nedd4-2 pathway, suggesting that RCTR1 may have potential as a therapeutic drug for ARDS/ALI (100). Ouabain has been reported to suppress the activation of the NF-κB and MAPK signaling pathways and attenuate lipopolysaccharide (LPS)-induced ALI (101). Additionally, lipoxin A4 activates alveolar epithelial Na,K-ATPase and enhances AFC (102). The electroporation-mediated transfer of Na,K-ATPase/ENaC plasmids has been revealed to improve lung function, reduce histopathological damage, decrease lung edema and increase survival in a translational porcine model of ARDS (103). Furthermore, pretreatment with cationic lipid-mediated transfer of Na,K-ATPase in vivo markedly reduces pulmonary edema by increasing Na,K-ATPase expression and activity (104).
Research suggests that the presence of the Na,K-ATPase α1 subunit attenuates Lyn (a member of the Src family of kinases) activation by LPS, which subsequently restricts downstream ROS and NF-κB signaling (105). Findings from β1 subunit knockout mouse model experiments demonstrated that AFC was markedly decreased (106), indicating a key role for the Na+ pump β1 subunit in alveolar ion and fluid transport. Claudin 18 is specifically expressed in lung epithelial cells and claudin 18 knockout mice present increased levels of AFC associated with elevated β-adrenergic receptor signaling, together with increased ENaC and Na,K-ATPase activity and increased Na,K-ATPase β1 subunit expression (107). Na,K-ATPase β1 subunit gene therapy not only upregulates tight junctions to ameliorate lung injury but also reduces lung permeability (108). Na,K-ATPase is a powerful target for ARDS/ALI treatment that is necessary for proper alveolar epithelial function because it promotes both barrier integrity and the resolution of excess alveolar fluid, thus enabling appropriate gas exchange.
CF
CF is an autosomal recessive disease caused by mutations in the gene encoding the CFTR, which leads to abnormal transport of ions and fluids across epithelial cell membranes, resulting in impaired mucociliary clearance (109–111). Respiratory failure is the most common cause of mortality in individuals with CF, leading to death in 90% of patients. Cystic fibrosis affects ~70,000 individuals worldwide. Although CF affects individuals of all ethnicities, it is most common in non-Hispanic White individuals (112). The majority of individuals with CF take inhaled medications daily to thin their mucus and use mechanical devices several times daily to dislodge mucus from the airways. Antibiotics may be prescribed to help control infection (113). Although there is an improvement in the management of CF for patients after the approval of CFTR modulators, this is still unsatisfactory, which includes a) non-significant response in F508del mutation heterozygotes to ivacaftor; b) requirement to continue other daily symptomatic treatment; c) interaction with CYP3A inducers and inhibitors and side effects such as elevated transaminases, cataract, oropharyngeal pain; d) negligible benefit in individuals aged <12 years old; and e) need of higher dose in case of lumacaftor and ivacaftor.
Multiple intra- and extracellular signaling pathways, such as HIF-1α, NF-κB, PI3K, AKT, and MAPKs, have been implicated in CF lung pathogenesis (114). Evidence indicates that inhibition of the PI3K/Akt/mTOR pathway improves CFTR stability and suggests that this pathway merits further study as a therapeutic target in CF (115). It has been reported that the activity of Na,K-ATPase in the lower airway epithelium is two-fold greater in patients with patients than in healthy controls. The increase in Na,K-ATPase activity, which contributes to increased reabsorption in epithelial cells and a compensatory mechanism of Na homeostasis, may carry out an important role in the pathogenesis of CF (116,117). Research has revealed that increased FXYD5 expression increases Na,K-ATPase activity, leading to increased transepithelial Na+ absorption in CF airway epithelial cells, which may further promote the development of CF (118). Among patients with CF, female patients have worse survival compared with male patients (119). Estrogen carries out a key role in the pathogenesis of CF by increasing Na absorption through the activation of Na,K-ATPase (120). The activation of Na,K-ATPase, which increases transepithelial Na+ absorption, results in mucociliary clearance dysfunction and ultimately promotes the progression of CF. Inhibition of Na,K-ATPase may help control the development of CF (121).
Idiopathic pulmonary fibrosis (IPF)
IPF is a progressive and fatal lung disease with unknown causes, characterized by progressive lung scarring and excessive extracellular matrix deposition, with limited available effective therapies (122). It mainly occurs in elderly adults with a median survival time of 2–3 years (123). Chronic microlesions in the alveolar epithelium and the accumulation of activated fibroblasts are the main pathophysiological processes associated with IPF. A portion of injured alveolar epithelial cells express mesenchymal markers and secrete profibrotic cytokines.
The Na,K-ATPase β1 subunit has been reported to be involved in organ fibrosis. Research has demonstrated the downregulation of Na,K-ATPase α1 and β1 subunits in the lung tissues of a bleomycin-induced lung fibrosis mouse model, as well as in TGF-β1-treated A549 cells (124,125). In a further study, Na,K-ATPase β1 subunit-deficient A549 cells exhibited increased expression of mesenchymal markers (126). Existing evidence suggested that overexpression of α-smooth muscle actin (α-SMA) in lung fibrosis was associated with the activation of PI3K/AKT12, and the interaction between TGF-β and PI3K/AKT promoted the formation of PF. Therefore, a potential approach to treating IPF may be inhibiting the PI3K/AKT/mTOR pathway (127).
In addition, ouabain has been revealed to have antifibrotic effects. In vitro, ouabain suppresses TGF-β1 by blocking the TGF-β/Smad signaling pathway and downregulating TGF-RII (128,129). It also ameliorates bleomycin-induced PF (130). An increase in the intracellular [Na+]/[K+] ratio drives the induction of cyclooxygenase-2 expression and PKA activation, which is accompanied by decreased Rho activation and myofibroblast differentiation in response to TGF-β (131,132). While ouabain may be a promising drug for the treatment of IPF, its narrow therapeutic window must be considered. Overall, Na,K-ATPase carries out an important role in IPF, with decreased expression in alveolar epithelial cells observed both in vitro and in vivo. Therefore, we hypothesize that ion dysfunction may be involved in the development of IPF.
Pulmonary hypertension (PH)
PH is a heterogeneous clinical disease characterized primarily by an abnormal increase in pulmonary artery pressure and is associated with adverse vascular remodeling with obstruction, stiffening and vasoconstriction of the pulmonary vasculature (133,134). At least 1% of the world's population is affected, with a greater burden more likely in low-income and middle-income countries (135). Five main groups of pulmonary hypertension are recognized: Pulmonary arterial hypertension (rare), pulmonary hypertension associated with left-sided heart disease (very common), pulmonary hypertension associated with lung disease (common), pulmonary hypertension associated with thromboembolic disease (rare), and pulmonary hypertension with unclear and/or multifactorial mechanisms (rare) (136). The current therapy primarily targets the underlying cause (137).
Research has demonstrated that FXYD1 protects against vascular dysfunction by preventing glutathionylation-induced oxidative inhibition of endothelial nitric oxide synthase (eNOS) in isolated human endothelial cells and in vascular tissue from mice (138). eNOS also increases Na,K-ATPase activity in a FXYD1-dependent manner (139). In further studies, FXYD1 knockout mice displayed characteristics of PH, including marked increases in pulmonary arterial pressure and increased muscularization of small pulmonary arterioles (140). In addition, ouabain-induced inhibition of Na+/K+-ATPase increases both Na+ and Ca2+ levels in the caveolae of the pulmonary artery smooth muscle plasma membrane, which could be important for the manifestation of PH (141). In conclusion, Na+/K+-ATPase carries out an important role in modulating redox and inflammatory signaling pathways involved in the development of PH and may serve as a potential therapeutic target for PH treatment.
Conclusion
Na,K-ATPase is known to act as an ion pump to modulate ion metabolism, however its role in signal transduction is less well understood. The present review briefly described the structure and function of Na,K-ATPase and discussed its role in lung diseases. Na,K-ATPase is involved in the development of various diseases and carries out a key role in their pathogenesis. Targeting Na,K-ATPase for the treatment of lung diseases may provide a novel therapeutic strategy. We hypothesize that it holds potential in the treatment of patients with heart failure complicated by pulmonary edema. However, further clinical evidence is necessary to substantiate its efficacy. Ouabain, a natural compound belonging to the cardiac glycoside family, is a Na,K-ATPase specific inhibitor with a systemic bioavailability of 43–50% after oral administration, and has anti-aging, anti-tumor, anti-fibrosis and improved cardiac function (142,143). Na,K-ATPase is also a neuroendocrine hormone synthesized in the adrenal cortex and hypothalamus (122–132,144). Moreover, hypoxia is an important stimulus for the release of endogenous ouabain (145). This relationship may draw more attention to the connection between Na,K-ATPase and lung diseases.
Acknowledgements
Not applicable.
Funding
Funding: No funding was received.
Availability of data and materials
Not applicable.
Authors' contributions
ZL and TD wrote the manuscript, BL and WZ edited and revised the manuscript, FL and CH interpreted the mechanism of action of Na, K-ATPase, prepared the figures and revised the manuscript. All authors read and approved the final manuscript. Data authentication not applicable.
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
Skou JC: The influence of some cations on an adenosine triphosphatase from peripheral nerves. Biochim Biophys Acta. 23:394–401. 1957. View Article : Google Scholar : PubMed/NCBI | |
Kaplan JH: Biochemistry of Na,K-ATPase. Annu Rev Biochem. 71:511–535. 2002. View Article : Google Scholar : PubMed/NCBI | |
Mobasheri A, Avila J, Cózar-Castellano I, Brownleader MD, Trevan M, Francis MJ, Lamb JF and Martín-Vasallo P: Na+, K+-ATPase isozyme diversity; comparative biochemistry and physiological implications of novel functional interactions. Biosci Rep. 20:51–91. 2000. View Article : Google Scholar : PubMed/NCBI | |
Rajasekaran SA, Gopal J, Willis D, Espineda C, Twiss JL and Rajasekaran AK: Na,K-ATPase beta1-subunit increases the translation efficiency of the alpha1-subunit in MSV-MDCK cells. Mol Biol Cell. 15:3224–3232. 2004. View Article : Google Scholar : PubMed/NCBI | |
Blanco G, Sánchez G and Mercer RW: Differential regulation of Na,K-ATPase isozymes by protein kinases and arachidonic acid. Arch Biochem Biophys. 359:139–150. 1998. View Article : Google Scholar : PubMed/NCBI | |
Benarroch EE: Na+, K+-ATPase: Functions in the nervous system and involvement in neurologic disease. Neurology. 76:287–293. 2011. View Article : Google Scholar : PubMed/NCBI | |
Lemas MV, Hamrick M, Takeyasu K and Fambrough DM: 26 Amino acids of an extracellular domain of the Na,K-ATPase alpha-subunit are sufficient for assembly with the Na,K-ATPase beta-subunit. J Biol Chem. 269:8255–8259. 1994. View Article : Google Scholar : PubMed/NCBI | |
Geering K, Meyer DI, Paccolat MP, Kraehenbühl JP and Rossier BC: Membrane insertion of alpha- and beta-subunits of Na+,K+-ATPase. J Biol Chem. 260:5154–5160. 1985. View Article : Google Scholar : PubMed/NCBI | |
Blanco G: Na,K-ATPase subunit heterogeneity as a mechanism for tissue-specific ion regulation. Semin Nephrol. 25:292–303. 2005. View Article : Google Scholar : PubMed/NCBI | |
Geering K: Functional roles of Na,K-ATPase subunits. Curr Opin Nephrol Hypertens. 17:526–532. 2008. View Article : Google Scholar : PubMed/NCBI | |
Geering K: FXYD proteins: new regulators of Na-K-ATPase. Am J Physiol Renal Physiol. 290:F241–F250. 2006. View Article : Google Scholar : PubMed/NCBI | |
Contreras RG, Torres-Carrillo A, Flores-Maldonado C, Shoshani L and Ponce A: Na+/K+-ATPase: More than an electrogenic pump. Int J Mol Sci. 25:61222024. View Article : Google Scholar : PubMed/NCBI | |
Laursen M, Gregersen JL, Yatime L, Nissen P and Fedosova UN: Structures and characterization of digoxin- and bufalin-bound Na+,K+-ATPase compared with the ouabain-bound complex. Proc Natl Acad Sci USA. 112:1755–1760. 2015. View Article : Google Scholar : PubMed/NCBI | |
Quintas LE, Pierre SV, Liu LJ, Bai Y, Liu XC and Xie ZJ: Alterations of Na+/K+-ATPase function in caveolin-1 knockout cardiac fibroblasts. J Mol Cell Cardiol. 49:525–531. 2010. View Article : Google Scholar : PubMed/NCBI | |
Skou JC: The identification of the sodium pump. Biosci Rep. 24:436–451. 2004. View Article : Google Scholar : PubMed/NCBI | |
Xie ZJ and Askari A: Na(+)/K(+)-ATPase as a signal transducer. Eur J Biochem. 269:2434–2439. 2002. View Article : Google Scholar : PubMed/NCBI | |
Pierre SV and Xie Z: The Na,K-ATPase receptor complex: Its organization and membership. Cell Biochem Biophys. 46:303–316. 2006. View Article : Google Scholar : PubMed/NCBI | |
Rajasekaran SA and Rajasekaran AK: Na,K-ATPase and epithelial tight junctions. Front Biosci (Landmark Ed). 14:2130–2148. 2009. View Article : Google Scholar : PubMed/NCBI | |
Barwe SP, Anikmar G, Moon SY, Zeng Y, Whitelegge JP, Rajasekaran SA and Rajasekaran AK: Novel role for Na,K-ATPase in phosphatidylinositol 3-kinase signaling and suppression of cell motility. Mol Biol Cell. 16:1082–1094. 2005. View Article : Google Scholar : PubMed/NCBI | |
Kimura T, Han W, Pagel P, Nairn AC and Caplan MJ: Protein phosphatase 2A interacts with the Na,K-ATPase and modulates its trafficking by inhibition of its association with arrestin. PLoS One. 6:e292692011. View Article : Google Scholar : PubMed/NCBI | |
Ren Y, Anderson AT, Meyer G, Lauber KM, Gallucci JC and Douglas Kinghorn A: Digoxin and its Na+/K+-ATPase-targeted actions on cardiovascular diseases and cancer. Bioorg Med Chem. 114:1179392024. View Article : Google Scholar : PubMed/NCBI | |
Maxwell KD, Chuang J, Chaudhry M, Nie Y, Bai F, Sodhi K, Liu J and Shapiro JI: The potential role of Na-K-ATPase and its signaling in the development of anemia in chronic kidney disease. Am J Physiol Renal Physiol. 320:F234–F242. 2021. View Article : Google Scholar : PubMed/NCBI | |
Bartlett DE, Miller RB, Thiesfeldt S, Lakhani HV, Shapiro JI and Sodhi K: The role of Na/K-ATPase signaling in oxidative stress related to aging: Implications in obesity and cardiovascular disease. Int J Mol Sci. 19:21392018. View Article : Google Scholar : PubMed/NCBI | |
Thai AA, Solomon BJ, Sequist LV, Gainor JF and Heist RE: Lung cancer. Lancet. 398:535–554. 2021. View Article : Google Scholar : PubMed/NCBI | |
Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ Jr, Wu YL and Paz-Ares L: Lung cancer: Current therapies and new targeted treatments. Lancet. 389:299–311. 2017. View Article : Google Scholar : PubMed/NCBI | |
Luo G, Zhang Y, Rumgay H, Morgan E, Langselius O, Vignat J, Colombet M and Bray F: Estimated worldwide variation and trends in incidence of lung cancer by histological subtype in 2022 and over time: A population-based study. Lancet Respir Med. 13:348–363. 2025. View Article : Google Scholar : PubMed/NCBI | |
Hirsch FR, Suda K, Wiens J and Bunn PA Jr: New and emerging targeted treatments in advanced non-small-cell lung cancer. Lancet. 388:1012–1024. 2016. View Article : Google Scholar : PubMed/NCBI | |
Stenkvist B, Bengtssion E, Dahlqvist B, Eriksson O, Jarkrans T and Nordin B: Cardiac glycosides and breast cancer, revisited. N Engl J Med. 306:4841982. View Article : Google Scholar : PubMed/NCBI | |
Siegel RL, Giaquinto AN and Jemal A: Cancer statistics, 2024. CA Cancer J Clin. 74:12–49. 2024.PubMed/NCBI | |
Felippe Gonçalves-de-Albuquerque C, Ribeiro Silva A, Ignácio da Silva C, Caire Castro-Faria-Neto H and Burth P: Na/K pump and beyond: Na/K-ATPase as a modulator of apoptosis and autophagy. Molecules. 22:5782017. View Article : Google Scholar : PubMed/NCBI | |
Bejček J, Spiwok V, Kmoníčková E and Rimpelová S: Na+/K+-ATPase revisited: On its mechanism of action, role in cancer, and activity modulation. Molecules. 26:19052021. View Article : Google Scholar : PubMed/NCBI | |
Diederich M, Muller F and Cerella C: Cardiac glycosides: From molecular targets to immunogenic cell death. Biochem Pharmacol. 125:1–11. 2017. View Article : Google Scholar : PubMed/NCBI | |
Huynh TP, Mah V, Sampson VB, Chia D, Fishbein MC, Horvath S, Alavi M, Wu DC, Harper J, Sarafian T, et al: Na,K-ATPase is a target of cigarette smoke and reduced expression predicts poor patient outcome of smokers with lung cancer. Am J Physiol Lung Cell Mol Physiol. 302:L1150–L1158. 2012. View Article : Google Scholar : PubMed/NCBI | |
Schneider C, Spaink H, Alexe G, Dharia NV, Meyer A, Merickel LA, Khalid D, Scheich S, Häupl B, Staudt LM, et al: Targeting the sodium-potassium pump as a therapeutic strategy in acute myeloid leukemia. Cancer Res. 84:3354–3370. 2024. View Article : Google Scholar : PubMed/NCBI | |
Espineda C, Seligson DB, Ball WJ Jr, Rao JY, Palotie A, Horvath S, Huang Y, Shi T and Rajasekaran AK: Analysis of the Na,K-ATPase alpha- and beta-subunit expression profiles of bladder cancer using tissue microarrays. Cancer. 97:1859–1868. 2003. View Article : Google Scholar : PubMed/NCBI | |
Rajasekaran SA, Ball WJ Jr, Bander NH, Liu H, Pardee JD and Rajasekaran AK: Reduced expression of beta-subunit of Na,K-ATPase in human clear-cell renal cell carcinoma. J Urol. 162:574–580. 1999. View Article : Google Scholar : PubMed/NCBI | |
Rajasekaran SA, Huynh TP, Wolle DG, Espineda CE, Inge LJ, Skay A, Lassman C, Nicholas SB, Harper JF, Reeves AE, et al: Na,K-ATPase subunits as markers for epithelial-mesenchymal transition in cancer and fibrosis. Mol Cancer Ther. 9:1515–1524. 2010. View Article : Google Scholar : PubMed/NCBI | |
Rajasekaran SA, Palmer LG, Moon SY, Soler AP, Apodaca GL, Harper JF, Zheng Y and Rajasekaran AK: Na,K-ATPase activity is required for formation of tight junctions, desmosomes, and induction of polarity in epithelial cells. Mol Biol Cell. 12:3717–3732. 2001. View Article : Google Scholar : PubMed/NCBI | |
Barwe SP, Kim S, Rajasekaran SA, Bowie JU and Rajasekaran KA: Janus model of the Na,K-ATPase beta-subunit transmembrane domain: Distinct faces mediate alpha/beta assembly and beta-beta homo-oligomerization. J Mol Biol. 365:706–714. 2007. View Article : Google Scholar : PubMed/NCBI | |
Inge LJ, Rajasekaran SA, Yoshimoto K, Mischel PS, McBride W, Landaw E and Rajasekaran KA: Evidence for a potential tumor suppressor role for the Na,K-ATPase beta1-subunit. Histol Histopathol. 23:459–467. 2008.PubMed/NCBI | |
Shoshani L, Contreras RG, Roldán ML, Moreno J, Lázaro A, Balda MS, Matter K and Cereijido M: The polarized expression of Na+,K+-ATPase in epithelia depends on the association between beta-subunits located in neighboring cells. Mol Biol Cell. 16:1071–1081. 2005. View Article : Google Scholar : PubMed/NCBI | |
Vagin O, Tokhtaeva E and Sachs G: The role of the beta1 subunit of the Na,K-ATPase and its glycosylation in cell-cell adhesion. J Biol Chem. 281:39573–39587. 2006. View Article : Google Scholar : PubMed/NCBI | |
Udoh UaS, 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 | |
Mijatovic T, Roland I, Quaquebeke EV, Nilsson B, Mathieu A, Vynckt FV, Darro F, Blanco G, Facchini V and Kiss R: The alpha1 subunit of the sodium pump could represent a novel target to combat non-small cell lung cancers. J Pathol. 212:170–179. 2007. View Article : Google Scholar : PubMed/NCBI | |
Liu CC, Kim YJ, Teh R, Garcia A, Hamilton EJ, Cornelius F, Baxter RC and Rasmussen HH: Displacement of native FXYD protein from Na+/K+-ATPase with novel FXYD peptide derivatives: Effects on doxorubicin cytotoxicity. Front Oncol. 12:8592162022. View Article : Google Scholar : PubMed/NCBI | |
Liu CC, Teh R, Mozar CA, Baxter RC and Rasmussen HH: Silencing overexpression of FXYD3 protein in breast cancer cells amplifies effects of doxorubicin and γ-radiation on Na(+)/K(+)-ATPase and cell survival. Breast Cancer Res Treat. 155:203–213. 2016. View Article : Google Scholar : PubMed/NCBI | |
Cordeiro BM, Leite Fontes CF and Meyer-Fernandes JR: Molecular basis of Na, K-ATPase regulation of diseases: Hormone and FXYD2 interactions. Int J Mol Sci. 25:133982024. View Article : Google Scholar : PubMed/NCBI | |
Okudela K, Yazawa T, Ishii J, Woo T, Mitsui H, Bunai T, Sakaeda M, Shimoyamada H, Sato H, Tajiri M, et al: Down-regulation of FXYD3 expression in human lung cancers: Its mechanism and potential role in carcinogenesis. Am J Pathol. 175:2646–2656. 2009. View Article : Google Scholar : PubMed/NCBI | |
Liu J, Feng Y, Zeng X, He M, Gong Y and Liu Y: Extracellular vesicles-encapsulated let-7i shed from bone mesenchymal stem cells suppress lung cancer via KDM3A/DCLK1/FXYD3 axis. J Cell Mol Med. 25:1911–1926. 2021. View Article : Google Scholar : PubMed/NCBI | |
Crambert G, Li C, Claeys D and Geering K: FXYD3 (Mat-8), a new regulator of Na,K-ATPase. Mol Biol Cell. 16:2363–2371. 2005. View Article : Google Scholar : PubMed/NCBI | |
Glaviano A, Foo ASC, Lam HY, Yap KCH, Jacot W, Jones RH, Eng H, Nair MG, Makvandi P, Geoerger B, et al: PI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancer. Mol Cancer. 22:1382023. View Article : Google Scholar : PubMed/NCBI | |
LoPiccolo J, Blumenthal GM, Bernstein WB and Dennis PA: Targeting the PI3K/Akt/mTOR pathway: Effective combinations and clinical considerations. Drug Resist Updat. 11:32–50. 2008. View Article : Google Scholar : PubMed/NCBI | |
Ren Y, Wu S, Burdette JE, Cheng X and Kinghorn AD: Structural insights into the interactions of digoxin and Na+/K+-ATPase and other targets for the inhibition of cancer cell proliferation. Molecules. 26:36722021. View Article : Google Scholar : PubMed/NCBI | |
Zhu Z, Sun H, Ma G, Wang Z, Li E and Liu Y and Liu Y: Bufalin induces lung cancer cell apoptosis via the inhibition of PI3K/Akt pathway. Int J Mol Sci. 13:2025–2035. 2012. View Article : Google Scholar : PubMed/NCBI | |
Chanvorachote P and Pongrakhananon V: Ouabain downregulates Mcl-1 and sensitizes lung cancer cells to TRAIL-induced apoptosis. Am J Physiol Cell Physiol. 304:C263–C272. 2013. View Article : Google Scholar : PubMed/NCBI | |
Yang K, Li Z, Chen Y, Yin F, Ji X, Zhou J, Li X, Zeng T, Fei C, Ren C, et al: Na, K-ATPase α1 cooperates with its endogenous ligand to reprogram immune microenvironment of lung carcinoma and promotes immune escape. Sci Adv. 9:eade53932023. View Article : Google Scholar : PubMed/NCBI | |
Song X, Xie L, Wang X, Zeng Q, Chen TC, Wang W and Song X: Temozolomide-perillyl alcohol conjugate induced reactive oxygen species accumulation contributes to its cytotoxicity against non-small cell lung cancer. Sci Rep. 6:227622016. View Article : Google Scholar : PubMed/NCBI | |
Lauf PK, Alqahtani T, Flues K, Meller L and Adragna NC: Interaction between Na-K-ATPase and Bcl-2 proteins BclXL and Bak. Am J Physiol Cell Physiol. 308:C51–C60. 2015. View Article : Google Scholar : PubMed/NCBI | |
Sun J, Chen L, Jiang P, Duan B, Wang R, Xu J, Liu W, Xu Y, Xie Z, Feng F and Qu W: Phenylethanoid glycosides of Callicarpa kwangtungensis Chun exert cardioprotective effect by weakening Na+-K+-ATPase/Src/ERK1/2 pathway and inhibiting apoptosis mediated by oxidative stress and inflammation. J Ethnopharmacol. 258:1128812020. View Article : Google Scholar : PubMed/NCBI | |
Wang Z, Pei S, Cui H, Zhang J and Jia Z: Zoonotic spillover and extreme weather events drive the global outbreaks of airborne viral emerging infectious diseases. J Med Virol. 96:e297372024. View Article : Google Scholar : PubMed/NCBI | |
Majumder J and Minko T: Recent developments on therapeutic and diagnostic approaches for COVID-19. AAPS J. 23:142021. View Article : Google Scholar : PubMed/NCBI | |
Ochani R, Asad A, Yasmin F, Shaikh S, Khalid H, Batra S, Sohail MR, Mahmood SF, Ochani R, Hussham Arshad M, et al: COVID-19 pandemic: From origins to outcomes. A comprehensive review of viral pathogenesis, clinical manifestations, diagnostic evaluation, and management. Infez Med. 29:20–36. 2021.PubMed/NCBI | |
Nyachoti DO, Fwelo P, Springer AE and Kelder SH: Association between gross national income per capita and COVID-19 vaccination coverage: A global ecological study. BMC Public Health. 23:24152023. View Article : Google Scholar : PubMed/NCBI | |
de Wit E, van Doremalen N, Falzarano D and Munster VJ: SARS and MERS: Recent insights into emerging coronaviruses. Nat Rev Microbiol. 14:523–534. 2016. View Article : Google Scholar : PubMed/NCBI | |
Guan WJ, Ni ZY, Hu Y, Liang WH, Ou CQ, He JX, Liu L, Shan H, Lei CL, Hui DSC, et al: Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 382:1708–1720. 2020. View Article : Google Scholar : PubMed/NCBI | |
Vitalii K and István V: Molecular mechanisms of Na,K-ATPase dysregulation driving alveolar epithelial barrier failure in severe COVID-19. Am J Physiol Lung Cell Mol Physiol. 320:L1186–L1193. 2021. View Article : Google Scholar : PubMed/NCBI | |
Souza E, Souza KFC, Moraes BPT, Paixão ICNP, Burth P, Silva AR and Gonçalves-de-Albuquerque CF: Na+/K+-ATPase as a target of cardiac glycosides for the treatment of SARS-CoV-2 infection. Front Pharmacol. 12:6247042021. View Article : Google Scholar : PubMed/NCBI | |
Blanco-Melo D, Nilsson-Payant BE, Liu WC, Uhl S, Hoagland D, Møller R, Jordan TX, Oishi K, Panis M, Sachs D, et al: Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell. 181:1036–1045.e9. 2020. View Article : Google Scholar : PubMed/NCBI | |
Chen F, Zhang Y, Sucgang R, Ramani S, Corry D, Kheradmand F and Creighton CJ: Meta-analysis of host transcriptional responses to SARS-CoV-2 infection reveals their manifestation in human tumors. Sci Rep. 11:24592021. View Article : Google Scholar : PubMed/NCBI | |
Ambade V and Ambade S: SARS-CoV-2 infecting endothelial cells, biochemical alterations, autopsy findings and outcomes in COVID-19, suggest role of hypoxia-inducible factor-1. J Med Biochem. 41:14–20. 2022. View Article : Google Scholar : PubMed/NCBI | |
Hamming I, Timens W, Bulthuis MLC, Lely AT, Navis GJ and van Goor H: Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol. 203:631–637. 2004. View Article : Google Scholar : PubMed/NCBI | |
Xie ZJ, Novograd J, Itzkowitz Y, Sher A, Buchen YD, Sodhi K, Abraham NG and Shapiro JI: The pivotal role of adipocyte-Na K peptide in reversing systemic inflammation in obesity and COVID-19 in the development of heart failure. Antioxidants (Basel). 9:11292020. View Article : Google Scholar : PubMed/NCBI | |
Li F, Li J, Wang PH, Yang N, Huang J, Ou J, Xu T, Zhao X, Liu T, Huang X, et al: SARS-CoV-2 spike promotes inflammation and apoptosis through autophagy by ROS-suppressed PI3K/AKT/mTOR signaling. Biochim Biophys Acta Mol Basis Dis. 1867:1662602021. View Article : Google Scholar : PubMed/NCBI | |
Basile MS, Cavalli E, McCubrey J, Hernández-Bello J, Muñoz-Valle JF, Fagone P and Nicoletti F: The PI3K/Akt/mTOR pathway: A potential pharmacological target in COVID-19. Drug Discov Today. 27:848–856. 2022. View Article : Google Scholar : PubMed/NCBI | |
Semenza GL: Hypoxia-inducible factors in physiology and medicine. Cell. 148:399–408. 2012. View Article : Google Scholar : PubMed/NCBI | |
Zhang H, Qian DZ, Tan YS, Lee K, Gao P, Ren YR, Rey S, Hammers H, Chang D, Pili R, et al: Digoxin and other cardiac glycosides inhibit HIF-1alpha synthesis and block tumor growth. Proc Natl Acad Sci USA. 105:19579–19586. 2008. View Article : Google Scholar : PubMed/NCBI | |
Ha DP, Shin WJ, Liu Z, Doche MiE, Lau R, Leli NM, Conn CS, Russo M, Lorenzato A, Koumenis C, et al: Targeting stress induction of GRP78 by cardiac glycoside oleandrin dually suppresses cancer and COVID-19. Cell Biosci. 14:1152024. View Article : Google Scholar : PubMed/NCBI | |
Newman RA, Sastry KJ, Arav-Boger R, Cai H, Matos R and Harrod R: Antiviral effects of oleandrin. J Exp Pharmacol. 12:503–515. 2020. View Article : Google Scholar : PubMed/NCBI | |
Van Kanegan MJ, Dunn DE, Kaltenbach LS, Shah B, He DN, McCoy DD, Yang PY, Peng JN, Shen L, Du L, et al: Dual activities of the anti-cancer drug candidate PBI-05204 provide neuroprotection in brain slice models for neurodegenerative diseases and stroke. Sci Rep. 6:256262016. View Article : Google Scholar : PubMed/NCBI | |
RECOVERY Collaborative Group and Horby P, ; Lim WS, Emberson JR, Mafham M, Bell JL, Linsell L, Staplin N, Brightling C, Ustianowski A, et al: Dexamethasone in hospitalized patients with Covid-19-preliminary report. N Engl J Med. 384:693–704. 2021. View Article : Google Scholar : PubMed/NCBI | |
Devarajan P and Benz EJ Jr: Translational regulation of Na-K-ATPase subunit mRNAs by glucocorticoids. Am J Physiol Renal Physiol. 279:F1132–F1138. 2000. View Article : Google Scholar : PubMed/NCBI | |
Matthay MA, Arabi Y, Arroliga AC, Bernard G, Bersten AD, Brochard LJ, Calfee CS, Combes A, Daniel BM, Ferguson ND, et al: A new global definition of acute respiratory distress syndrome. Am J Respir Crit Care Med. 209:37–47. 2024. View Article : Google Scholar : PubMed/NCBI | |
Matthay MA, Zemans RL, Zimmerman GA, Arabi YM, Beitler JR, Mercat A, Herridge M, Randolph AG and Calfee CS: Acute respiratory distress syndrome. Nat Rev Dis Primers. 5:182019. View Article : Google Scholar : PubMed/NCBI | |
Bernard GR, Artigas A, Brigham KL, Carlet J, Falke K, Hudson L, Lamy M, LeGall JR, Morris A and Spragg R: Report of the American-European consensus conference on ARDS: Definitions, mechanisms, relevant outcomes and clinical trial coordination. The consensus committee. Intensive Care Med. 20:225–232. 1994. View Article : Google Scholar : PubMed/NCBI | |
ARDS Definition Task Force, . Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND, Caldwell E, Fan E, Camporota L and Slutsky AS: Acute respiratory distress syndrome: The Berlin definition. JAMA. 307:2526–2533. 2012.PubMed/NCBI | |
Hu Q, Zhang S, Yang Y, Yao JQ, Tang WF, Lyon CJ, Hu TY and Wan MH: Extracellular vesicles in the pathogenesis and treatment of acute lung injury. Mil Med Res. 9:612022.PubMed/NCBI | |
Meyer NJ, Gattinoni L and Calfee CS: Acute respiratory distress syndrome. Lancet. 398:622–637. 2021. View Article : Google Scholar : PubMed/NCBI | |
Herold S, Gabrielli NM and Vadász I: Novel concepts of acute lung injury and alveolar-capillary barrier dysfunction. Am J Physiol Lung Cell Mol Physiol. 305:L665–L681. 2013. View Article : Google Scholar : PubMed/NCBI | |
Matthay MA, Folkesson HG and Clerici C: Lung epithelial fluid transport and the resolution of pulmonary edema. Physiol Rev. 82:569–600. 2002. View Article : Google Scholar : PubMed/NCBI | |
Gusarova GA, Trejo HE, Dada LA, Briva A, Welch LC, Hamanaka RB, Mutlu GM, Chandel NS, Prakriya M and Sznajder JI: Hypoxia leads to Na,K-ATPase downregulation via Ca(2+) release-activated Ca(2+) channels and AMPK activation. Mol Cell Biol. 31:3546–3556. 2011. View Article : Google Scholar : PubMed/NCBI | |
Berger G, Guetta J, Klorin G, Badarneh R, Braun E, Brod V, Saleh NA, Katz A, Bitterman H and Azzam ZS: Sepsis impairs alveolar epithelial function by downregulating Na-K-ATPase pump. Am J Physiol Lung Cell Mol Physiol. 301:L23–L30. 2011. View Article : Google Scholar : PubMed/NCBI | |
Peteranderl C, Morales-Nebreda L, Selvakumar B, Lecuona E, Vadász I, Morty RE, Schmoldt C, Bespalowa J, Wolff T, Pleschka S, et al: Macrophage-epithelial paracrine crosstalk inhibits lung edema clearance during influenza infection. J Clin Invest. 126:1566–1580. 2016. View Article : Google Scholar : PubMed/NCBI | |
Gonçalves-de-Albuquerque CF, Silva AR, Burth P, Castro-Faria MV and Castro-Faria-Neto HC: Acute Respiratory Distress Syndrome: Role of oleic acid-triggered lung injury and inflammation. Mediators Inflamm. 2015:2604652015. View Article : Google Scholar : PubMed/NCBI | |
Gonçalves-de-Albuquerque CF, Burth P, Silva AR, de Moraes IM, de Jesus Oliveira FM, Santelli RE, Freire AS, Bozza PT, Younes-Ibrahim M and de Castro-Faria MV: Oleic acid inhibits lung Na/K-ATPase in mice and induces injury with lipid body formation in leukocytes and eicosanoid production. J Inflamm (Lond). 10:342013. View Article : Google Scholar : PubMed/NCBI | |
Silva AR, de Souza E Souza KFC, Souza TB, Younes-Ibrahim M, Burth P, de Castro Faria Neto HC and Gonçalves-de-Albuquerque CF: The Na/K-ATPase role as a signal transducer in lung inflammation. Front Immunol. 14:12875122024. View Article : Google Scholar : PubMed/NCBI | |
Lei J and Ingbar DH: Src kinase integrates PI3K/Akt and MAPK/ERK1/2 pathways in T3-induced Na-K-ATPase activity in adult rat alveolar cells. Am J Physiol Lung Cell Mol Physiol. 301:L765–L771. 2011. View Article : Google Scholar : PubMed/NCBI | |
Upadhyay D, Lecuona E, Comellas A, Kamp DW and Sznajder JI: Fibroblast growth factor-10 upregulates Na,K-ATPase via the MAPK pathway. FEBS Lett. 545:173–176. 2003. View Article : Google Scholar : PubMed/NCBI | |
Wang Q, Yan SF, Hao Y and Jin SW: Specialized pro-resolving mediators regulate alveolar fluid clearance during acute respiratory distress syndrome. Chin Med J (Engl). 131:982–989. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zhang JL, Zhuo XJ, Lin J, Luo LC, Ying WY, Xie X, Zhang HW, Yang JX, Li D, Smith FG and Jin SW: Maresin1 stimulates alveolar fluid clearance through the alveolar epithelial sodium channel Na,K-ATPase via the ALX/PI3K/Nedd4-2 pathway. Lab Invest. 97:543–554. 2017. View Article : Google Scholar : PubMed/NCBI | |
Yang Q, Xu HR, Xiang SY, Zhang C, Ye Y, Shen CX, Mei HX, Zhang PH, Ma HY, Zheng SX, et al: Resolvin conjugates in tissue regeneration 1 promote alveolar fluid clearance by activating alveolar epithelial sodium channels and Na, K-ATPase in lipopolysaccharide-induced acute lung injury. J Pharmacol Exp Ther. 379:156–165. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wang C, Meng Y, Wang Y, Jiang Z, Xu M, Bo L and Deng X: Ouabain protects mice against lipopolysaccharide-induced acute lung injury. Med Sci Monit. 24:4455–4464. 2018. View Article : Google Scholar : PubMed/NCBI | |
Wang Q, Lian QQ, Li R, Ying BY, He Q, Chen F, Zheng X, Yang Y, Wu DR, Zheng SX, et al: Lipoxin A(4) activates alveolar epithelial sodium channel, Na,K-ATPase, and increases alveolar fluid clearance. Am J Respir Cell Mol Biol. 48:610–618. 2013. View Article : Google Scholar : PubMed/NCBI | |
Emr BM, Roy S, Kollisch-Singule M, Gatto LA, Barravecchia M, Lin X, Young JL, Wang G, Liu J, Satalin J, et al: Electroporation-mediated gene delivery of Na+,K+-ATPase, and ENaC subunits to the lung attenuates acute respiratory distress syndrome in a two-hit porcine model. Shock. 43:16–23. 2015. View Article : Google Scholar : PubMed/NCBI | |
Stern M, Ulrich K, Robinson C, Copeland J, Griesenbach U, Masse C, Cheng S, Munkonge F, Geddes D, Berthiaume Y and Alton E: Pretreatment with cationic lipid-mediated transfer of the Na+K+-ATPase pump in a mouse model in vivo augments resolution of high permeability pulmonary edema. Gene Ther. 7:960–966. 2000. View Article : Google Scholar : PubMed/NCBI | |
Zhang J, Chang J, Beg MA, Huang W, Zhao Y, Dai W, Wu X, Cui W, Pillai SS, Lakhani HV, et al: Na/K-ATPase suppresses LPS-induced pro-inflammatory signaling through Lyn. iScience. 25:1049632022. View Article : Google Scholar : PubMed/NCBI | |
Flodby P, Kim YH, Beard LL, Gao D, Ji Y, Kage H, Liebler JM, Minoo P, Kim KJ, Borok Z and Crandall ED: Knockout mice reveal a major role for alveolar epithelial type I cells in alveolar fluid clearance. Am J Respir Cell Mol Biol. 55:395–406. 2016. View Article : Google Scholar : PubMed/NCBI | |
Li G, Flodby P, Luo J, Kage H, Sipos A, Gao D, Ji Y, Beard LL, Marconett CN, DeMaio L, et al: Knockout mice reveal key roles for claudin 18 in alveolar barrier properties and fluid homeostasis. Am J Respir Cell Mol Biol. 51:210–222. 2014. View Article : Google Scholar : PubMed/NCBI | |
Lin X, Barravecchia M, Kothari P, Young JL and Dean DA: β1-Na(+),K(+)-ATPase gene therapy upregulates tight junctions to rescue lipopolysaccharide-induced acute lung injury. Gene Ther. 23:489–499. 2016. View Article : Google Scholar : PubMed/NCBI | |
Kerem B, Rommens JM, Buchanan JA, Markiewicz D, Cox TK, Chakravarti A, Buchwald M and Tsui LC: Identification of the cystic fibrosis gene: Genetic analysis. Science. 245:1073–1080. 1989. View Article : Google Scholar : PubMed/NCBI | |
Riordan JR, Romments JM, Kerem B, Alon N, Rozmahel R, Grzelczak Z, Zielenski J, Lok S, Plavsic N, Chou JL, et al: Identification of the cystic fibrosis gene: Cloning and characterization of complementary DNA. Science. 245:1066–1073. 1989. View Article : Google Scholar : PubMed/NCBI | |
Rommens JM, Iannuzzi MC, Kerem B, Drumm ML, Melmer G, Dean M, Rozmahel R, Cole JL, Kennedy D, Hidaka N, et al: Identification of the cystic fibrosis gene: Chromosome walking and jumping. Science. 245:1059–1065. 1989. View Article : Google Scholar : PubMed/NCBI | |
Endres TM and Konstan MW: What is cystic fibrosis? JAMA. 327:1912022. View Article : Google Scholar : PubMed/NCBI | |
Rafeeq MM and Murad HAS: Cystic fibrosis: Current therapeutic targets and future approaches. J Transl Med. 15:842017. View Article : Google Scholar : PubMed/NCBI | |
Natarajan V: Is PI3K a villain in cystic fibrosis? Am J Respir Cell Mol Biol. 62:552–553. 2020. View Article : Google Scholar : PubMed/NCBI | |
Reilly R, Mroz MS, Dempsey E, Wynne K, Keely SJ, McKone EF, Hiebel C, Behl C and Coppinger JA: Targeting the PI3K/Akt/mTOR signalling pathway in cystic fibrosis. Sci Rep. 7:76422017. View Article : Google Scholar : PubMed/NCBI | |
Peckham D, Holland E, Range S and Knox AJ: Na+/K+ ATPase in lower airway epithelium from cystic fibrosis and non-cystic-fibrosis lung. Biochem Biophys Res Commun. 232:464–468. 1997. View Article : Google Scholar : PubMed/NCBI | |
Luczay A, Vásárhelyi B, Dobos M, Holics K, Ujhelyi R and Tulassay T: Altered erythrocyte sodium-lithium counter-transport and Na+/K(+)-ATPase activity in cystic fibrosis. Acta Paediatr. 86:245–247. 1997. View Article : Google Scholar : PubMed/NCBI | |
Miller TJ and Davis PB: FXYD5 modulates Na+ absorption and is increased in cystic fibrosis airway epithelia. Am J Physiol Lung Cell Mol Physiol. 294:L654–L664. 2008. View Article : Google Scholar : PubMed/NCBI | |
Raghavan D, Gao A, Ahn C, Kaza V, Finklea J, Torres F and Jain R: Lung transplantation and gender effects on survival of recipients with cystic fibrosis. J Heart Lung Transplant. 35:1487–1496. 2016. View Article : Google Scholar : PubMed/NCBI | |
Saint-Criq V, Kim SH, Katzenellenbogen JA and Harvey BJ: Non-genomic estrogen regulation of ion transport and airway surface liquid dynamics in cystic fibrosis bronchial epithelium. PLoS One. 8:e785932013. View Article : Google Scholar : PubMed/NCBI | |
Matalon S, Bartoszewski R and Collawn JF: Role of epithelial sodium channels in the regulation of lung fluid homeostasis. Am J Physiol Lung Cell Mol Physiol. 309:L1229–L1238. 2015. View Article : Google Scholar : PubMed/NCBI | |
Richeldi L, Collard HR and Jones MG: Idiopathic pulmonary fibrosis. Lancet. 389:1941–1952. 2017. View Article : Google Scholar : PubMed/NCBI | |
Cai M, Zhu M, Ban C, Su J, Ye Q, Liu Y, Zhao W, Wang C and Dai H: Clinical features and outcomes of 210 patients with idiopathic pulmonary fibrosis. Chin Med J (Engl). 127:1868–1873. 2014. View Article : Google Scholar : PubMed/NCBI | |
Li B, Huang X, Xu X, Ning W, Dai H and Wang C: The profibrotic effect of downregulated Na,K-ATPase β1 subunit in alveolar epithelial cells during lung fibrosis. Int J Mol Med. 44:273–280. 2019.PubMed/NCBI | |
Rodrigo R, Trujillo S, Bosco C, Orellana M, Thielemann L and Araya J: Changes in (Na + K)-adenosine triphosphatase activity and ultrastructure of lung and kidney associated with oxidative stress induced by acute ethanol intoxication. Chest. 121:589–596. 2002. View Article : Google Scholar : PubMed/NCBI | |
Nam LB and Keum YS: Regulation of NRF2 by Na+/K+-ATPase: Implication of tyrosine phosphorylation of Src. Free Radic Res. 54:883–893. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wang J, Hu K, Cai X, Yang B, He Q, Wang J and Weng Q: Targeting PI3K/AKT signaling for treatment of idiopathic pulmonary fibrosis. Acta Pharm Sin B. 12:18–32. 2022. View Article : Google Scholar : PubMed/NCBI | |
Ludens JH, Clark MA, DuCharme DW, Harris DW, Lutzke BS, Mandel F, Mathews WR, Sutter DM and Hamlyn JM: Purification of an endogenous digitalislike factor from human plasma for structural analysis. Hypertension. 17:923–929. 1991. View Article : Google Scholar : PubMed/NCBI | |
Sophocleous A, Elmatzoglou I and Souvatzoglou A: Circulating endogenous digitalis-like factor(s) (EDLF) in man is derived from the adrenals and its secretion is ACTH-dependent. J Endocrinol Invest. 26:668–674. 2003. View Article : Google Scholar : PubMed/NCBI | |
Li B, Huang X, Liu Z, Xu X, Xiao H, Zhang X, Dai H and Wang C: Ouabain ameliorates bleomycin induced pulmonary fibrosis by inhibiting proliferation and promoting apoptosis of lung fibroblasts. Am J Transl Res. 10:2967–2974. 2018.PubMed/NCBI | |
La J, Reed EB, Koltsova S, Akimova O, Hamanaka RB, Mutlu GM, Orlov SN and Dulin NO: Regulation of myofibroblast differentiation by cardiac glycosides. Am J Physiol Lung Cell Mol Physiol. 310:L815–L823. 2016. View Article : Google Scholar : PubMed/NCBI | |
Orlov SN, La J, Smolyaninova LV and Dulin NO: Na+,K+-ATPase as a target for treatment of tissue fibrosis. Curr Med Chem. 26:564–575. 2019. View Article : Google Scholar : PubMed/NCBI | |
Johnson S, Sommer N, Cox-Flaherty K, Weissmann N, Ventetuolo CE and Maron BA: Pulmonary hypertension: A contemporary review. Am J Respir Crit Care Med. 208:528–548. 2023. View Article : Google Scholar : PubMed/NCBI | |
Mocumbi A, Humbert M, Saxena A, Jing ZC, Sliwa K, Thienemann F, Archer SL and Stewart S: Pulmonary hypertension. Nat Rev Dis Primers. 10:12024. View Article : Google Scholar : PubMed/NCBI | |
Humbert M, Kovacs G, Hoeper MM, Badagliacca R, Berger RMF, Brida M, Carlsen J, Coats AJS, Escribano-Subias P, Ferrari P, et al: 2022 ESC/ERS guidelines for the diagnosis and treatment of pulmonary hypertension. Eur Respir J. 61:22008792023. View Article : Google Scholar : PubMed/NCBI | |
Kovacs G, Bartolome S, Denton CP, Gatzoulis MA, Gu S, Khanna D, Badesch D and Montani D: Definition, classification and diagnosis of pulmonary hypertension. Eur Respir J. 64:24013242024. View Article : Google Scholar : PubMed/NCBI | |
Poch D and Mandel J: Pulmonary hypertension. Ann Intern Med. 174:ITC49–ITC64. 2021. View Article : Google Scholar : PubMed/NCBI | |
Bubb KJ, Tang O, Gentile C, Moosavi SM, Hansen T, Liu CC, Di Bartolo BA and Figtree GA: FXYD1 is protective against vascular dysfunction. Hypertension. 77:2104–2116. 2021. View Article : Google Scholar : PubMed/NCBI | |
Pavlovic D, Hall AR, Kennington EJ, Aughton K, Boguslavskyi A, Fuller W, Despa S, Bers DM and Shattock MJ: Nitric oxide regulates cardiac intracellular Na+ and Ca2+by modulating Na/K ATPase via PKCε and phospholemman-dependent mechanism. J Mol Cell Cardiol. 61:164–171. 2013. View Article : Google Scholar : PubMed/NCBI | |
Hansen TS, Karimi Galougahi K, Tang O, Tsang M, Scherrer-Crosbie M, Arystarkhova E, Sweadner K, Bursill C, Bubb KJ and Figtree GA: The FXYD1 protein plays a protective role against pulmonary hypertension and arterial remodeling via redox and inflammatory mechanisms. Am J Physiol Heart Circ Physiol. 326:H623–H635. 2024. View Article : Google Scholar : PubMed/NCBI | |
Ghosh B, Kar P, Mandal A, Dey K, Chakraborti T and Chakraborti S: Ca2+ influx mechanisms in caveolae vesicles of pulmonary smooth muscle plasma membrane under inhibition of alpha2beta1 isozyme of Na+/K+-ATPase by ouabain. Life Sci. 84:139–148. 2009. View Article : Google Scholar : PubMed/NCBI | |
Fürstenwerth H: Ouabain-the insulin of the heart. Int J Clin Pract. 64:1591–1594. 2010. View Article : Google Scholar : PubMed/NCBI | |
Guerrero A, Herranz N, Sun B, Wagner V, Gallage S, Guiho R, Wolter K, Pombo J, Irvine EE, Innes AJ, et al: Cardiac glycosides are broad-spectrum senolytics. Nat Metab. 1:1074–1088. 2019. View Article : Google Scholar : PubMed/NCBI | |
Laredo J, Hamilton BP and Hamlyn JM: Secretion of endogenous ouabain from bovine adrenocortical cells: Role of the zona glomerulosa and zona fasciculata. Biochem Biophys Res Commun. 212:487–493. 1995. View Article : Google Scholar : PubMed/NCBI | |
De Angelis C and Haupert GT Jr: Hypoxia triggers release of an endogenous inhibitor of Na(+)-K(+)-ATPase from midbrain and adrenal. Am J Physiol. 274:F182–F188. 1998.PubMed/NCBI |