1
|
Blaustein MP, Hamlyn JM. Sensational site: the sodium pump ouabain-binding site and its ligands. Am J Physiol Cell Physiol 2024; 326:C1120-C1177. [PMID: 38223926 PMCID: PMC11193536 DOI: 10.1152/ajpcell.00273.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/22/2023] [Accepted: 01/10/2024] [Indexed: 01/16/2024]
Abstract
Cardiotonic steroids (CTS), used by certain insects, toads, and rats for protection from predators, became, thanks to Withering's trailblazing 1785 monograph, the mainstay of heart failure (HF) therapy. In the 1950s and 1960s, we learned that the CTS receptor was part of the sodium pump (NKA) and that the Na+/Ca2+ exchanger was critical for the acute cardiotonic effect of digoxin- and ouabain-related CTS. This "settled" view was upended by seven revolutionary observations. First, subnanomolar ouabain sometimes stimulates NKA while higher concentrations are invariably inhibitory. Second, endogenous ouabain (EO) was discovered in the human circulation. Third, in the DIG clinical trial, digoxin only marginally improved outcomes in patients with HF. Fourth, cloning of NKA in 1985 revealed multiple NKA α and β subunit isoforms that, in the rodent, differ in their sensitivities to CTS. Fifth, the NKA is a cation pump and a hormone receptor/signal transducer. EO binding to NKA activates, in a ligand- and cell-specific manner, several protein kinase and Ca2+-dependent signaling cascades that have widespread physiological effects and can contribute to hypertension and HF pathogenesis. Sixth, all CTS are not equivalent, e.g., ouabain induces hypertension in rodents while digoxin is antihypertensinogenic ("biased signaling"). Seventh, most common rodent hypertension models require a highly ouabain-sensitive α2 NKA and the elevated blood pressure is alleviated by EO immunoneutralization. These numerous phenomena are enabled by NKA's intricate structure. We have just begun to understand the endocrine role of the endogenous ligands and the broad impact of the ouabain-binding site on physiology and pathophysiology.
Collapse
Affiliation(s)
- Mordecai P Blaustein
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - John M Hamlyn
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
2
|
Staehr C, Aalkjaer C, Matchkov V. The vascular Na,K-ATPase: clinical implications in stroke, migraine, and hypertension. Clin Sci (Lond) 2023; 137:1595-1618. [PMID: 37877226 PMCID: PMC10600256 DOI: 10.1042/cs20220796] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 10/26/2023]
Abstract
In the vascular wall, the Na,K-ATPase plays an important role in the control of arterial tone. Through cSrc signaling, it contributes to the modulation of Ca2+ sensitivity in vascular smooth muscle cells. This review focuses on the potential implication of Na,K-ATPase-dependent intracellular signaling pathways in severe vascular disorders; ischemic stroke, familial migraine, and arterial hypertension. We propose similarity in the detrimental Na,K-ATPase-dependent signaling seen in these pathological conditions. The review includes a retrospective proteomics analysis investigating temporal changes after ischemic stroke. The analysis revealed that the expression of Na,K-ATPase α isoforms is down-regulated in the days and weeks following reperfusion, while downstream Na,K-ATPase-dependent cSrc kinase is up-regulated. These results are important since previous studies have linked the Na,K-ATPase-dependent cSrc signaling to futile recanalization and vasospasm after stroke. The review also explores a link between the Na,K-ATPase and migraine with aura, as reduced expression or pharmacological inhibition of the Na,K-ATPase leads to cSrc kinase signaling up-regulation and cerebral hypoperfusion. The review discusses the role of an endogenous cardiotonic steroid-like compound, ouabain, which binds to the Na,K-ATPase and initiates the intracellular cSrc signaling, in the pathophysiology of arterial hypertension. Currently, our understanding of the precise control mechanisms governing the Na,K-ATPase/cSrc kinase regulation in the vascular wall is limited. Understanding the role of vascular Na,K-ATPase signaling is essential for developing targeted treatments for cerebrovascular disorders and hypertension, as the Na,K-ATPase is implicated in the pathogenesis of these conditions and may contribute to their comorbidity.
Collapse
Affiliation(s)
- Christian Staehr
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 35, Aarhus, Denmark
| | - Christian Aalkjaer
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
- Danish Cardiovascular Academy, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
| | - Vladimir V. Matchkov
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
| |
Collapse
|
3
|
Rognant S, Kravtsova VV, Bouzinova EV, Melnikova EV, Krivoi II, Pierre SV, Aalkjaer C, Jepps TA, Matchkov VV. The microtubule network enables Src kinase interaction with the Na,K-ATPase to generate Ca2+ flashes in smooth muscle cells. Front Physiol 2022; 13:1007340. [PMID: 36213229 PMCID: PMC9538378 DOI: 10.3389/fphys.2022.1007340] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 09/05/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Several local Ca2+ events are characterized in smooth muscle cells. We have previously shown that an inhibitor of the Na,K-ATPase, ouabain induces spatially restricted intracellular Ca2+ transients near the plasma membrane, and suggested the importance of this signaling for regulation of intercellular coupling and smooth muscle cell contraction. The mechanism behind these Na,K-ATPase-dependent “Ca2+ flashes” remains to be elucidated. In addition to its conventional ion transport function, the Na,K-ATPase is proposed to contribute to intracellular pathways, including Src kinase activation. The microtubule network is important for intracellular signaling, but its role in the Na,K-ATPase-Src kinase interaction is not known. We hypothesized the microtubule network was responsible for maintaining the Na,K-ATPase-Src kinase interaction, which enables Ca2+ flashes. Methods: We characterized Ca2+ flashes in cultured smooth muscle cells, A7r5, and freshly isolated smooth muscle cells from rat mesenteric artery. Cells were loaded with Ca2+-sensitive fluorescent dyes, Calcium Green-1/AM and Fura Red/AM, for ratiometric measurements of intracellular Ca2+. The Na,K-ATPase α2 isoform was knocked down with siRNA and the microtubule network was disrupted with nocodazole. An involvement of the Src signaling was tested pharmacologically and with Western blot. Protein interactions were validated with proximity ligation assays. Results: The Ca2+ flashes were induced by micromolar concentrations of ouabain. Knockdown of the α2 isoform Na,K-ATPase abolished Ca2+ flashes, as did inhibition of tyrosine phosphorylation with genistein and PP2, and the inhibitor of the Na,K-ATPase-dependent Src activation, pNaKtide. Ouabain-induced Ca2+ flashes were associated with Src kinase activation by phosphorylation. The α2 isoform Na,K-ATPase and Src kinase colocalized in the cells. Disruption of microtubule with nocodazole inhibited Ca2+ flashes, reduced Na,K-ATPase/Src interaction and Src activation. Conclusion: We demonstrate that the Na,K-ATPase-dependent Ca2+ flashes in smooth muscle cells require an interaction between the α2 isoform Na, K-ATPase and Src kinase, which is maintained by the microtubule network.
Collapse
Affiliation(s)
- Salomé Rognant
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Violetta V. Kravtsova
- Department of General Physiology, St. Petersburg State University, St. Petersburg, Russia
| | | | | | - Igor I. Krivoi
- Department of General Physiology, St. Petersburg State University, St. Petersburg, Russia
| | - Sandrine V. Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV, United States
| | | | - Thomas A. Jepps
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vladimir V. Matchkov
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- *Correspondence: Vladimir V. Matchkov,
| |
Collapse
|
4
|
Cai J, Zhang BD, Li YQ, Zhu WF, Akihisa T, Kikuchi T, Xu J, Liu WY, Feng F, Zhang J. Cardiac glycosides from the roots of Streblus asper Lour. with activity against Epstein-Barr virus lytic replication. Bioorg Chem 2022; 127:106004. [PMID: 35843015 DOI: 10.1016/j.bioorg.2022.106004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 11/15/2022]
Abstract
Cardiac glycosides (CGs) show potential broad-spectrum antiviral activity by targeting cellular host proteins. Herein are reported the isolation of five new (1-5) and eight known (7-13) CGs from the roots of Streblus asper Lour. Of these compounds 1 and 7 exhibited inhibitory action against EBV early antigen (EA) expression, with half-maximal effective concentration values (EC50) being less than 60 nM, and they also showed selectivity, with selectivity index (SI) values being 56.80 and 103.17, respectively. Preliminary structure activity relationships indicated that the C-10 substituent, C-5 hydroxy groups, and C-3 sugar unit play essential roles in the mediation of the inhibitory activity of CGs against EBV. Further enzyme experiments demonstrated that these compounds might inhibit ion pump function and thereby change the intracellular signal transduction pathway by binding to Na+/K+-ATPase, as validated by simulated molecular docking. This study is the first report that CGs can effectively limit EBV lytic replication, and the observations made in this study may be of value for lead compound development.
Collapse
Affiliation(s)
- Jing Cai
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Bo-Dou Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Yu-Qi Li
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Wan-Fang Zhu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China; School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Toshihiro Akihisa
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China; Research Institute for Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Takashi Kikuchi
- Faculty of Pharmaceutical Sciences, Toho University, Chiba 274-8510, Japan
| | - Jian Xu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Wen-Yuan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Feng Feng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China; Jiangsu Food and Pharmaceutical Science College, Huaian 223003, China
| | - Jie Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China; Jiangsu Food and Pharmaceutical Science College, Huaian 223003, China.
| |
Collapse
|
5
|
Shandell MA, Capatina AL, Lawrence SM, Brackenbury WJ, Lagos D. Inhibition of the Na +/K +-ATPase by cardiac glycosides suppresses expression of the IDO1 immune checkpoint in cancer cells by reducing STAT1 activation. J Biol Chem 2022; 298:101707. [PMID: 35150740 PMCID: PMC8902613 DOI: 10.1016/j.jbc.2022.101707] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/18/2022] Open
Abstract
Despite extensive basic and clinical research on immune checkpoint regulatory pathways, little is known about the effects of the ionic tumor microenvironment on immune checkpoint expression and function. Here we describe a mechanistic link between Na+/K+-ATPase (NKA) inhibition and activity of the immune checkpoint protein indoleamine-pyrrole 2',3'-dioxygenase 1 (IDO1). We found that IDO1 was necessary and sufficient for production of kynurenine, a downstream tryptophan metabolite, in cancer cells. We developed a spectrophotometric assay to screen a library of 31 model ion transport-targeting compounds for potential effects on IDO1 function in A549 lung and MDA-MB-231 breast cancer cells. This revealed that the cardiac glycosides ouabain and digoxin inhibited kynurenine production at concentrations that did not affect cell survival. NKA inhibition by ouabain and digoxin resulted in increased intracellular Na+ levels and downregulation of IDO1 mRNA and protein levels, which was consistent with the reduction in kynurenine levels. Knockdown of ATP1A1, the ɑ1 subunit of the NKA and target of cardiac glycosides, increased Na+ levels to a lesser extent than cardiac glycoside treatment and did not affect IDO1 expression. However, ATP1A1 knockdown significantly enhanced the effect of cardiac glycosides on IDO1 expression and kynurenine production. Mechanistically, we show that cardiac glycoside treatment resulted in curtailing the length of phosphorylation-mediated stabilization of STAT1, a transcriptional regulator of IDO1 expression, an effect enhanced by ATP1A1 knockdown. Our findings reveal cross talk between ionic modulation via cardiac glycosides and immune checkpoint protein expression in cancer cells with broad mechanistic and clinical implications.
Collapse
Affiliation(s)
- Mia A Shandell
- Department of Biology, University of York, York, United Kingdom; Hull York Medical School, University of York, York, United Kingdom; York Biomedical Research Institute, University of York, York, United Kingdom
| | - Alina L Capatina
- Department of Biology, University of York, York, United Kingdom; York Biomedical Research Institute, University of York, York, United Kingdom
| | | | - William J Brackenbury
- Department of Biology, University of York, York, United Kingdom; York Biomedical Research Institute, University of York, York, United Kingdom
| | - Dimitris Lagos
- Hull York Medical School, University of York, York, United Kingdom; York Biomedical Research Institute, University of York, York, United Kingdom.
| |
Collapse
|
6
|
Mirkamali M, Momeni HR, Etemadi T, Mosayebi G, Komijani M. Involvement of caspase-3 in apoptosis of human lymphocytes exposed to cadmium chloride. Hum Exp Toxicol 2022; 41:9603271221121796. [PMID: 36036252 DOI: 10.1177/09603271221121796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Lymphocytes are a group of white blood cells with a variety of roles their integrity is crucial for the body's immune responses. Cadmium, a heavy metal and environmental pollutant, is known as a toxicant to exert its adverse effects on some sort of cells including blood cells. RESEARCH DESIGN In this study, human lymphocytes were divided into 3 groups: (1) lymphocytes at 0-h, (2) lymphocytes at 24 h (control), (3) lymphocytes treated with cadmium chloride (15 μM). Lymphocyte viability and plasma membrane integrity were assessed in these groups. In addition, the occurrence of apoptosis was investigated by assessment of nucleus diameter and flow cytometry. Activation of caspase-3 was also detected by immunocytochemistry. RESULTS Result showed that lymphocyte's viability and plasma membrane integrity decreased in lymphocytes treated with cadmium as compared with the control group. Decreased nucleus diameter and result of flow cytometry demonstrated cadmium-induced apoptosis in human lymphocytes. Furthermore, lymphocytes treated with cadmium displayed intensely activated caspase-3 immunoreactivity in their cytoplasm. CONCLUSION In conclusion, cadmium not only negatively effect on viability and plasma membrane, but also induces caspase-dependent apoptosis in human lymphocytes.
Collapse
Affiliation(s)
- Mona Mirkamali
- Biology Department, Faculty of Science, 125649Arak University, Arak, Iran
| | - Hamid Reza Momeni
- Biology Department, Faculty of Science, 125649Arak University, Arak, Iran
| | - Tahereh Etemadi
- Biology Department, Faculty of Science, 125649Arak University, Arak, Iran
| | - Ghasem Mosayebi
- Department of Microbiology and Immunology, Molecular and Medicine Research Center (MMRC), 48412Arak University of Medical Sciences, Arak, Iran
| | - Majid Komijani
- Biology Department, Faculty of Science, 125649Arak University, Arak, Iran
| |
Collapse
|
7
|
Kidney Cancer and Chronic Kidney Disease: Too Close for Comfort. Biomedicines 2021; 9:biomedicines9121761. [PMID: 34944574 PMCID: PMC8699019 DOI: 10.3390/biomedicines9121761] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/18/2021] [Accepted: 11/21/2021] [Indexed: 12/11/2022] Open
Abstract
Kidney cancer and chronic kidney disease are two renal pathologies with very different clinical management strategies and therapeutical options. Nonetheless, the cellular and molecular mechanisms underlying both conditions are closely related. Renal physiology is adapted to operate with a limited oxygen supply, making the kidney remarkably equipped to respond to hypoxia. This tightly regulated response mechanism is at the heart of kidney cancer, leading to the onset of malignant cellular phenotypes. Although elusive, the role of hypoxia in chronic kidney diseases is emerging as related to fibrosis, a pivotal factor in decaying renal function. The present review offers a perspective on the common biological traits shared between kidney cancer and chronic kidney disease and the available and prospective therapies for both conditions.
Collapse
|
8
|
Digoxin exerts anticancer activity on human nonsmall cell lung cancer cells by blocking PI3K/Akt pathway. Biosci Rep 2021; 41:229832. [PMID: 34549269 PMCID: PMC8495431 DOI: 10.1042/bsr20211056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 01/03/2023] Open
Abstract
Lung cancer remains the leading cause of cancer mortality because of its metastatic potential and high malignancy. The discovery of new applications for old drugs is a shortcut for cancer therapy. We recently investigated the antitumor effect of digoxin, a well-established drug for treating heart failure, against nonsmall cell lung cancer A549 and H1299 cells. Digoxin inhibited the proliferation and colony-forming ability of the two cell lines and arrested the cell cycle at the G0/G1 phase in A549 cells and the G2/M phase in H1299 cells. Mitochondria-mediated apoptosis was induced in A549 cells but not in H1299 cells after treatment with digoxin. Moreover, digoxin inhibited the migration, invasion, adhesion and epithelial–mesenchymal transition of A549 and H1299 cells. Autophagy was induced in both cell lines after treatment with digoxin, with an increase in autophagosome foci. In addition, digoxin inhibited the phosphorylation of Akt, mTOR and p70S6K, signaling molecules of the PI3K/Akt pathway that are known to be involved in tumor cell survival, proliferation, metastasis and autophagy. Our findings suggest that digoxin has the potential to be used for therapy for human nonsmall cell lung cancer, but further evidence is required.
Collapse
|
9
|
Mohan S, Tiwari MN, Stanojević M, Biala Y, Yaari Y. Muscarinic regulation of the neuronal Na + /K + -ATPase in rat hippocampus. J Physiol 2021; 599:3735-3754. [PMID: 34148230 DOI: 10.1113/jp281460] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/16/2021] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Stimulation of postsynaptic muscarinic receptors was shown to excite principal hippocampal neurons by modulating several membrane ion conductances. We show here that activation of postsynaptic muscarinic receptors also causes neuronal excitation by inhibiting Na+ /K+ -ATPase activity. Muscarinic Na+ /K+ -ATPase inhibition is mediated by two separate signalling pathways that lead downstream to enhanced Na+ /K+ -ATPase phosphorylation by activating protein kinase C and protein kinase G. Muscarinic excitation through Na+ /K+ -ATPase inhibition is probably involved in cholinergic modulation of hippocampal activity and may turn out to be a widespread mechanism of neuronal excitation in the brain. ABSTRACT Stimulation of muscarinic cholinergic receptors on principal hippocampal neurons enhances intrinsic neuronal excitability by modulating several membrane ion conductances. The electrogenic Na+ /K+ -ATPase (NKA; the 'Na+ pump') is a ubiquitous regulator of intrinsic neuronal excitability, generating a hyperpolarizing current to thwart excessive neuronal firing. Using electrophysiological and pharmacological methodologies in rat hippocampal slices, we show that neuronal NKA pumping activity is also subjected to cholinergic regulation. Stimulation of postsynaptic muscarinic, but not nicotinic, cholinergic receptors activates membrane-bound phospholipase C and hydrolysis of membrane-integral phosphatidylinositol 4,5-bisphosphate into diacylglycerol (DAG) and inositol 1,4,5-triphosphate (IP3 ). Along one signalling pathway, DAG activates protein kinase C (PKC). Along a second signalling pathway, IP3 causes Ca2+ release from the endoplasmic reticulum, facilitating nitric oxide (NO) production. The rise in NO levels stimulates cGMP synthesis by guanylate-cyclase, activating protein kinase G (PKG). The two pathways converge to cause partial NKA inhibition through enzyme phosphorylation by PKC and PKG, leading to a marked increase in intrinsic neuronal excitability. This novel mechanism of neuronal NKA regulation probably contributes to the cholinergic modulation of hippocampal activity in spatial navigation, learning and memory.
Collapse
Affiliation(s)
- Sandesh Mohan
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah School of Medicine, Jerusalem, 91120, Israel
| | - Manindra Nath Tiwari
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah School of Medicine, Jerusalem, 91120, Israel
| | - Marija Stanojević
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah School of Medicine, Jerusalem, 91120, Israel
| | - Yoav Biala
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah School of Medicine, Jerusalem, 91120, Israel
| | - Yoel Yaari
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah School of Medicine, Jerusalem, 91120, Israel
| |
Collapse
|
10
|
Valvassori SS, Dal-Pont GC, Varela RB, Resende WR, Gava FF, Mina FG, Budni J, Quevedo J. Ouabain induces memory impairment and alter the BDNF signaling pathway in an animal model of bipolar disorder: Cognitive and neurochemical alterations in BD model. J Affect Disord 2021; 282:1195-1202. [PMID: 33601696 DOI: 10.1016/j.jad.2020.12.190] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/16/2020] [Accepted: 12/26/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The present study aims to evaluate the effects of ouabain on memory and neurotrophic parameters in the brains of rats. METHODS Wistar rats received an intracerebroventricular (ICV) injection of ouabain or artificial cerebrospinal fluid (aCSF). Seven and 14 days after ICV administration, the animals were subjected to the open-field and splash tests. Furthermore, the pro-BDNF, BDNF, TrkB, and CREB were assessed in the frontal cortex and hippocampus of the rats, in both seven and 14 days after ICV injection. The memory of the animals was tested by novel object recognition test (NOR) and inhibitory avoidance task (IA), only 14 days after ICV administration. RESULTS Ouabain increased locomotion and exploration in the animals seven days after its administration; however, 14 days after ICV, these behavioral parameters return to the basal level. Seven days after ouabain administration increased grooming behavior in the splash test; on the other hand, seven days after ouabain injection decreased the grooming behavior, which is considered an anhedonic response. Besides, ouabain decreased recognition index in the NOR and decreased aversive memory in the IA, when compared to the control group. The levels of pro-BDNF and BDNF decreased in the frontal cortex seven days after ouabain; but its receptor (TrkB) and CREB decreased seven and 14 days after ouabain, in both cerebral structures evaluated. CONCLUSION Ouabain-induced animal model of BD is an excellent model to assess memory alteration, observed in bipolar patients. Besides, the memory impairment induced by ouabain seems to be related to BDNF signaling pathway alterations.
Collapse
Affiliation(s)
- Samira S Valvassori
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| | - Gustavo C Dal-Pont
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Roger B Varela
- Queensland Brain Institute, The Universty of Queensland, St Lucia, QLD 4072, Australia
| | - Wilson R Resende
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Fernanda F Gava
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Franciele G Mina
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Josiane Budni
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil; Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States; Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States
| |
Collapse
|
11
|
Petrushanko IY, Mitkevich VA, Makarov AA. Molecular Mechanisms of the Redox Regulation of the Na,K-ATPase. Biophysics (Nagoya-shi) 2020. [DOI: 10.1134/s0006350920050139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
12
|
Reddy D, Kumavath R, Barh D, Azevedo V, Ghosh P. Anticancer and Antiviral Properties of Cardiac Glycosides: A Review to Explore the Mechanism of Actions. Molecules 2020; 25:E3596. [PMID: 32784680 PMCID: PMC7465415 DOI: 10.3390/molecules25163596] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/19/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiac glycosides (CGs) have a long history of treating cardiac diseases. However, recent reports have suggested that CGs also possess anticancer and antiviral activities. The primary mechanism of action of these anticancer agents is by suppressing the Na+/k+-ATPase by decreasing the intracellular K+ and increasing the Na+ and Ca2+. Additionally, CGs were known to act as inhibitors of IL8 production, DNA topoisomerase I and II, anoikis prevention and suppression of several target genes responsible for the inhibition of cancer cell proliferation. Moreover, CGs were reported to be effective against several DNA and RNA viral species such as influenza, human cytomegalovirus, herpes simplex virus, coronavirus, tick-borne encephalitis (TBE) virus and Ebola virus. CGs were reported to suppress the HIV-1 gene expression, viral protein translation and alters viral pre-mRNA splicing to inhibit the viral replication. To date, four CGs (Anvirzel, UNBS1450, PBI05204 and digoxin) were in clinical trials for their anticancer activity. This review encapsulates the current knowledge about CGs as anticancer and antiviral drugs in isolation and in combination with some other drugs to enhance their efficiency. Further studies of this class of biomolecules are necessary to determine their possible inhibitory role in cancer and viral diseases.
Collapse
Affiliation(s)
- Dhanasekhar Reddy
- Department of Genomic Science, School of Biological Sciences, University of Kerala, Tejaswini Hills, Periya (P.O), Kasaragod, Kerala 671320, India;
| | - Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences, University of Kerala, Tejaswini Hills, Periya (P.O), Kasaragod, Kerala 671320, India;
| | - Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur WB-721172, India;
| | - Vasco Azevedo
- Laboratório de Genética Celular e Molecular, Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal deMinas Gerais (UFMG), Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA;
| |
Collapse
|
13
|
The Na/K-ATPase α1 and c-Src form signaling complex under native condition: A crosslinking approach. Sci Rep 2020; 10:6006. [PMID: 32265464 PMCID: PMC7138855 DOI: 10.1038/s41598-020-61920-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 03/04/2020] [Indexed: 11/09/2022] Open
Abstract
The protein-protein interactions amongst the Na/K-ATPase α1 subunit, c-Src, and caveolin-1 (cav-1) are essential for the Na/K-ATPase signaling functions. However, there are arguments concerning the interaction model. The present study aims to clarify the interactions amongst the endogenous native proteins in live cells under native resting condition. Under native condition, Blue Native-PAGE and Blue Native-PAGE/SDS-PAGE 2D analyses demonstrated co-existence of the α1 subunit and c-Src in same protein complex, as well as a direct interaction between the α1 subunit and c-Src. By comparison of cleavable and non-cleavable cysteine-cysteine crosslinked samples, capillary immunoblotting analysis demonstrated that depletion of Src kinase family members (c-Src, Yes, and Fyn) or cav-1 clearly reduced the interactions of the α1 subunit with proteins, but depletion of cav-1 did not affect the interaction of c-Src with the α1 subunit. The data indicated that there are direct interactions between the α1 subunit and c-Src as well as between the α1 subunit and cav-1, but argued about the interaction between c-Src and cav-1 under the condition. Furthermore, the data also indicated the existence of different protein complexes containing the α1 subunit and c-Src, which might have different signaling functions.
Collapse
|
14
|
Dumas SJ, Meta E, Borri M, Goveia J, Rohlenova K, Conchinha NV, Falkenberg K, Teuwen LA, de Rooij L, Kalucka J, Chen R, Khan S, Taverna F, Lu W, Parys M, De Legher C, Vinckier S, Karakach TK, Schoonjans L, Lin L, Bolund L, Dewerchin M, Eelen G, Rabelink TJ, Li X, Luo Y, Carmeliet P. Single-Cell RNA Sequencing Reveals Renal Endothelium Heterogeneity and Metabolic Adaptation to Water Deprivation. J Am Soc Nephrol 2019; 31:118-138. [PMID: 31818909 DOI: 10.1681/asn.2019080832] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/01/2019] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Renal endothelial cells from glomerular, cortical, and medullary kidney compartments are exposed to different microenvironmental conditions and support specific kidney processes. However, the heterogeneous phenotypes of these cells remain incompletely inventoried. Osmotic homeostasis is vitally important for regulating cell volume and function, and in mammals, osmotic equilibrium is regulated through the countercurrent system in the renal medulla, where water exchange through endothelium occurs against an osmotic pressure gradient. Dehydration exposes medullary renal endothelial cells to extreme hyperosmolarity, and how these cells adapt to and survive in this hypertonic milieu is unknown. METHODS We inventoried renal endothelial cell heterogeneity by single-cell RNA sequencing >40,000 mouse renal endothelial cells, and studied transcriptome changes during osmotic adaptation upon water deprivation. We validated our findings by immunostaining and functionally by targeting oxidative phosphorylation in a hyperosmolarity model in vitro and in dehydrated mice in vivo. RESULTS We identified 24 renal endothelial cell phenotypes (of which eight were novel), highlighting extensive heterogeneity of these cells between and within the cortex, glomeruli, and medulla. In response to dehydration and hypertonicity, medullary renal endothelial cells upregulated the expression of genes involved in the hypoxia response, glycolysis, and-surprisingly-oxidative phosphorylation. Endothelial cells increased oxygen consumption when exposed to hyperosmolarity, whereas blocking oxidative phosphorylation compromised endothelial cell viability during hyperosmotic stress and impaired urine concentration during dehydration. CONCLUSIONS This study provides a high-resolution atlas of the renal endothelium and highlights extensive renal endothelial cell phenotypic heterogeneity, as well as a previously unrecognized role of oxidative phosphorylation in the metabolic adaptation of medullary renal endothelial cells to water deprivation.
Collapse
Affiliation(s)
- Sébastien J Dumas
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Elda Meta
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Mila Borri
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Jermaine Goveia
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Katerina Rohlenova
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Nadine V Conchinha
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Kim Falkenberg
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Laure-Anne Teuwen
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Laura de Rooij
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Joanna Kalucka
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Rongyuan Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Shawez Khan
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Federico Taverna
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Weisi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Magdalena Parys
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Carla De Legher
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Stefan Vinckier
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Tobias K Karakach
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Luc Schoonjans
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Lin Lin
- Lars Bolund Institute of Regenerative Medicine, Beijing Genomics Institute (BGI)-Qingdao, BGI-Shenzhen, Qingdao, China.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Lars Bolund
- Lars Bolund Institute of Regenerative Medicine, Beijing Genomics Institute (BGI)-Qingdao, BGI-Shenzhen, Qingdao, China.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Mieke Dewerchin
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Guy Eelen
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Ton J Rabelink
- Division of Nephrology, Department of Internal Medicine, The Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China;
| | - Yonglun Luo
- Lars Bolund Institute of Regenerative Medicine, Beijing Genomics Institute (BGI)-Qingdao, BGI-Shenzhen, Qingdao, China; .,Department of Biomedicine, Aarhus University, Aarhus, Denmark.,China National GeneBank, Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China; and.,Qingdao-Europe Advanced Institute for Life Sciences, Beijing Genomics Institute (BGI)-Qingdao, Qingdao, China
| | - Peter Carmeliet
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium; .,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
15
|
Askari A. The other functions of the sodium pump. Cell Calcium 2019; 84:102105. [DOI: 10.1016/j.ceca.2019.102105] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/21/2019] [Accepted: 10/28/2019] [Indexed: 01/14/2023]
|
16
|
Staehr C, Hangaard L, Bouzinova EV, Kim S, Rajanathan R, Boegh Jessen P, Luque N, Xie Z, Lykke-Hartmann K, Sandow SL, Aalkjaer C, Matchkov VV. Smooth muscle Ca 2+ sensitization causes hypercontractility of middle cerebral arteries in mice bearing the familial hemiplegic migraine type 2 associated mutation. J Cereb Blood Flow Metab 2019; 39. [PMID: 29513112 PMCID: PMC6681533 DOI: 10.1177/0271678x18761712] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Familial hemiplegic migraine type 2 (FHM2) is associated with inherited point-mutations in the Na,K-ATPase α2 isoform, including G301R mutation. We hypothesized that this mutation affects specific aspects of vascular function, and thus compared cerebral and systemic arteries from heterozygote mice bearing the G301R mutation (Atp1a2+/-G301R) with wild type (WT). Middle cerebral (MCA) and mesenteric small artery (MSA) function was compared in an isometric myograph. Cerebral blood flow was assessed with Laser speckle analysis. Intracellular Ca2+ and membrane potential were measured simultaneously. Protein expression was semi-quantified by immunohistochemistry. Protein phosphorylation was analysed by Western blot. MSA from Atp1a2+/-G301R and WT showed similar contractile responses. The Atp1a2+/-G301R MCA constricted stronger to U46619, endothelin and potassium compared to WT. This was associated with an increased depolarization, although the Ca2+ change was smaller than in WT. The enhanced constriction of Atp1a2+/-G301R MCA was associated with increased cSrc activation, stronger sensitization to [Ca2+]i and increased MYPT1 phosphorylation. These differences were abolished by cSrc inhibition. Atp1a2+/-G301R mice had reduced resting blood flow through MCA in comparison with WT mice. FHM2-associated mutation leads to elevated contractility of MCA due to sensitization of the contractile machinery to Ca2+, which is mediated via Na,K-ATPase/Src-kinase/MYPT1 signalling.
Collapse
Affiliation(s)
| | - Lise Hangaard
- 1 Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Sukhan Kim
- 1 Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | | - Nathan Luque
- 2 Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Queensland, Australia
| | - Zijian Xie
- 3 Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV, USA
| | | | - Shaun L Sandow
- 2 Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Queensland, Australia
| | | | | |
Collapse
|
17
|
Askari A. The sodium pump and digitalis drugs: Dogmas and fallacies. Pharmacol Res Perspect 2019; 7:e00505. [PMID: 31360524 PMCID: PMC6639696 DOI: 10.1002/prp2.505] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/20/2019] [Accepted: 05/24/2019] [Indexed: 11/29/2022] Open
Abstract
The sodium pump (Na/K-ATPase) is a plasma membrane enzyme that transports Na+ and K+ against their physiological gradients in most eukaryotic cells. Besides pumping ions, the enzyme may also interact with neighboring proteins to activate cell signaling pathways that regulate cell growth. Digitalis drugs, useful for the treatment of heart failure and atrial arrhythmias, inhibit the pumping function of Na/K-ATPase and stimulate its signaling function. In the current field of research on the sodium pump and digitalis drugs, some issues that are commonly accepted to be well established are not so, and this may impede progress. Here, several such issues are identified, their histories are discussed, and their open discussions are urged. The covered unsettled questions consist of (a) the suggested hormonal role of endogenous digitalis compounds; (b) the specificity of Na/K-ATPase as the receptor for digitalis compounds; (c) the relevance of the positive inotropic action of digitalis to its use for the treatment of heart failure; (d) the conflicting findings on digitalis-induced signaling function of Na/K-ATPase; and (e) the uncertainties about the structure of Na/K-ATPase in the native cell membrane.
Collapse
Affiliation(s)
- Amir Askari
- Department of Cancer Biology, College of Medicine and Life SciencesUniversity of ToledoToledoOhio
| |
Collapse
|
18
|
Serum albumin saturation test based on non-esterified fatty acids imbalance for clinical employment. Clin Chim Acta 2019; 495:422-428. [PMID: 31082361 DOI: 10.1016/j.cca.2019.05.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 04/15/2019] [Accepted: 05/04/2019] [Indexed: 01/13/2023]
Abstract
Fatty acids are fundamental as energy and structural source to the human cells. They are not usually found free in human circulation. Alteration in fatty acids metabolism is linked to diseases such as diabetes, preeclampsia, heart disease, and some infectious diseases. Increased levels of non-esterified fatty acids (NEFA) may cause cell dysfunction and lipotoxicity. Since physiologically fatty acids are transported bound to albumin, we propose here a simple and cheap test that consists of albumin isoelectric focusing determination to measure the potential systemic NEFA cytotoxicity. For validation of this method, albumin isoelectric focusing in 51 serum samples from 40 critically ill patients and 11 controls was compared with NEFA/albumin ratios measured by HPLC. We called this approach an albumin saturation test. This test may indicate to physicians the potential NEFA lipotoxicity guiding them throughout better patient management. The albumin saturation test can point out serum albumin-NEFA saturation through a cheap assay that could be performed by any care facility.
Collapse
|
19
|
Effect of photobiomodulation therapy on neuronal injuries by ouabain: the regulation of Na, K-ATPase; Src; and mitogen-activated protein kinase signaling pathway. BMC Neurosci 2019; 20:19. [PMID: 31027504 PMCID: PMC6486688 DOI: 10.1186/s12868-019-0499-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 04/08/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To determine whether photobiomodulation (PBM) rescued the disruption of Na+/Ca2+ homeostasis and mitochondrial membrane potential by ouabain; the Na, K-ATPase inhibitor. For PBM in this study, a 660 nm LED array was used at energy densities of 0.78, 1.56, 3.12, 6.24, and 9.36 J/cm2. RESULTS HCN-2 neuronal cells treated with ouabain showed loss of cell polarity, disrupted cell morphology, and decreased cell viability, which were improved after PBM treatment. We found that ouabain-induced Na, K-ATPase inhibition promoted activation of downstream signaling through Src, Ras, and mitogen-activated protein kinase (MAPK), which were suppressed after PBM treatment. This provided evidence of Na, K-ATPase α-subunit inactivation and intracellular Ca2+ increase. In response to ouabain, we observed activation of Src and MAPK by Na, K-ATPase, decreased mitochondrial membrane potential, and Na+-dependent Ca2+ increases, which were restored by PBM treatment. CONCLUSIONS This study demonstrated that Na+/K+ imbalance could be regulated by PBM treatment in neuronal cells, and we suggest that PBM is a potential therapeutic tool for Na, K-ATPase targeted neuronal diseases.
Collapse
|
20
|
Abstract
The Na,K-ATPase is an enzyme essential for ion homeostasis in all cells. Over the last decades, it has been well-established that in addition to the transport of Na+/K+ over the cell membrane, the Na,K-ATPase acts as a receptor transducing humoral signals intracellularly. It has been suggested that ouabain-like compounds serve as endogenous modulators of this Na,K-ATPase signal transduction. The molecular mechanisms underlying Na,K-ATPase signaling are complicated and suggest the confluence of divergent biological pathways. This review discusses recent updates on the Na,K-ATPase signaling pathways characterized or suggested in vascular smooth muscle cells. The conventional view on this signaling is based on a microdomain structure where the Na,K-ATPase controls the Na,Ca-exchanger activity via modulation of intracellular Na+ in the spatially restricted submembrane space. This, in turn, affects intracellular Ca2+ and Ca2+ load in the sarcoplasmic reticulum leading to modulation of contractility as well as gene expression. An ion-transport-independent signal transduction from the Na,K-ATPase is based on molecular interactions. This was primarily characterized in other cell types but recently also demonstrated in vascular smooth muscles. The downstream signaling from the Na,K-ATPase includes Src and phosphatidylinositol-4,5-bisphosphate 3 kinase signaling pathways and generation of reactive oxygen species. Moreover, in vascular smooth muscle cells the interaction between the Na,K-ATPase and proteins responsible for Ca2+ homeostasis, e.g., phospholipase C and inositol triphosphate receptors, contributes to an integration of the signaling pathways. Recent update on the Na,K-ATPase dependent intracellular signaling and the significance for physiological functions and pathophysiological changes are discussed in this review.
Collapse
|
21
|
The Na/K-ATPase Signaling: From Specific Ligands to General Reactive Oxygen Species. Int J Mol Sci 2018; 19:ijms19092600. [PMID: 30200500 PMCID: PMC6163532 DOI: 10.3390/ijms19092600] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/10/2018] [Accepted: 08/28/2018] [Indexed: 12/16/2022] Open
Abstract
The signaling function of the Na/K-ATPase has been established for 20 years and is widely accepted in the field, with many excellent reports and reviews not cited here. Even though there is debate about the underlying mechanism, the signaling function is unquestioned. This short review looks back at the evolution of Na/K-ATPase signaling, from stimulation by cardiotonic steroids (also known as digitalis-like substances) as specific ligands to stimulation by reactive oxygen species (ROS) in general. The interplay of cardiotonic steroids and ROS in Na/K-ATPase signaling forms a positive-feedback oxidant amplification loop that has been implicated in some pathophysiological conditions.
Collapse
|
22
|
Lan YL, Yu ZL, Lou JC, Ma XC, Zhang B. Update on the effects of the sodium pump α1 subunit on human glioblastoma: from the laboratory to the clinic. Expert Opin Investig Drugs 2018; 27:753-763. [PMID: 30130132 DOI: 10.1080/13543784.2018.1512582] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Glioblastoma is a debilitating disease that is associated with poor prognosis and a very limited response to therapies; thus, molecularly targeted therapeutics and personalized therapy are urgently needed. The Na+/K+-ATPase sodium pump is a transmembrane protein complex that has recently been recognized as an important transducer and integrator of various signals. The sodium pump α1 subunit, which is highly expressed in most glioblastomas compared with that in normal brain tissues, is an emerging cancer target that merits further investigation. AREAS COVERED The purpose of this narrative review is to explore the important roles of the sodium pump α1 subunit in glioblastoma and analyze its potential therapeutic applications. EXPERT OPINION Expression of the sodium pump α1 subunit in glioblastoma tissues is generally higher than that in normal tissues. Sodium pump α1 subunit-mediated pivotal antiglioblastoma signaling pathways have been reviewed, and their impact on the sensitivity of glioblastoma cells to anticancer drugs has recently been clarified. In addition, various pharmacologically optimized sodium pump inhibitors have recently reached early clinical trials, and explorations of sodium pump α1 subunit inhibitors may hold promise for the development of stratification strategies in which patients are treated based on their isoform expression status.
Collapse
Affiliation(s)
- Yu-Long Lan
- a Department of Neurosurgery , The Second Affiliated Hospital of Dalian Medical University , Dalian , China.,b Department of Pharmacy , Dalian Medical University , Dalian , China.,c Department of Physiology , Dalian Medical University , Dalian , China
| | - Zhen-Long Yu
- b Department of Pharmacy , Dalian Medical University , Dalian , China
| | - Jia-Cheng Lou
- a Department of Neurosurgery , The Second Affiliated Hospital of Dalian Medical University , Dalian , China
| | - Xiao-Chi Ma
- b Department of Pharmacy , Dalian Medical University , Dalian , China
| | - Bo Zhang
- a Department of Neurosurgery , The Second Affiliated Hospital of Dalian Medical University , Dalian , China
| |
Collapse
|
23
|
Dal-Pont GC, Resende WR, Varela RB, Menegas S, Trajano KS, Peterle BR, Quevedo J, Valvassori SS. Inhibition of GSK-3β on Behavioral Changes and Oxidative Stress in an Animal Model of Mania. Mol Neurobiol 2018; 56:2379-2393. [DOI: 10.1007/s12035-018-1226-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 07/05/2018] [Indexed: 12/27/2022]
|
24
|
Liu J, Lilly MN, Shapiro JI. Targeting Na/K-ATPase Signaling: A New Approach to Control Oxidative Stress. Curr Pharm Des 2018; 24:359-364. [PMID: 29318961 PMCID: PMC6052765 DOI: 10.2174/1381612824666180110101052] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 12/27/2017] [Accepted: 01/04/2017] [Indexed: 01/13/2023]
Abstract
Renal and cardiac function are greatly affected by chronic oxidative stress which can cause many pathophysiological states. The Na/K-ATPase is well-described as an ion pumping enzyme involved in maintaining cellular ion homeostasis; however, in the past two decades, extensive research has been done to understand the signaling function of the Na/K-ATPase and determine its role in physiological and pathophysiological states. Our lab has shown that the Na/K-ATPase signaling cascade can function as an amplifier of reactive oxygen species (ROS) which can be initiated by cardiotonic steroids or increases in ROS. Regulation of systemic oxidative stress by targeting Na/K-ATPase signaling mediated oxidant amplification improves 5/6th partial nephrectomy (PNx) mediated uremic cardiomyopathy, renal sodium handling, as well as ameliorates adipogenesis. This review will present this new concept of Na/K-ATPase signaling mediated oxidant amplification loop and its clinic implication.
Collapse
Affiliation(s)
- Jiang Liu
- Dept. of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | - Megan N. Lilly
- Dept. of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | - Joseph I. Shapiro
- Dept. of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| |
Collapse
|
25
|
Ouabain affects cell migration via Na,K-ATPase-p130cas and via nucleus-centrosome association. PLoS One 2017; 12:e0183343. [PMID: 28817661 PMCID: PMC5560699 DOI: 10.1371/journal.pone.0183343] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/02/2017] [Indexed: 01/12/2023] Open
Abstract
Na,K-ATPase is a membrane protein that catalyzes ATP to maintain transmembrane sodium and potassium gradients. In addition, Na,K-ATPase also acts as a signal-transducing receptor for cardiotonic steroids such as ouabain and activates a number of signalling pathways. Several studies report that ouabain affects cell migration. Here we used ouabain at concentrations far below those required to block Na,K-ATPase pump activity and show that it significantly reduced RPE cell migration through two mechanisms. It causes dephosphorylation of a 130 kD protein, which we identify as p130cas. Src is involved, because Src inhibitors, but not inhibitors of other kinases tested, caused a similar reduction in p130cas phosphorylation and ouabain increased the association of Na,K-ATPase and Src. Knockdown of p130cas by siRNA reduced cell migration. Unexpectedly, ouabain induced separation of nucleus and centrosome, also leading to a block in cell migration. Inhibitor and siRNA experiments show that this effect is mediated by ERK1,2. This is the first report showing that ouabain can regulate cell migration by affecting nucleus-centrosome association.
Collapse
|
26
|
Plössl K, Royer M, Bernklau S, Tavraz NN, Friedrich T, Wild J, Weber BHF, Friedrich U. Retinoschisin is linked to retinal Na/K-ATPase signaling and localization. Mol Biol Cell 2017; 28:2178-2189. [PMID: 28615319 PMCID: PMC5531734 DOI: 10.1091/mbc.e17-01-0064] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 01/30/2023] Open
Abstract
Retinoschisin binds to the extracellular domain of Na/K-ATPase subunit β2. Retinoschisin inhibits Na/K-ATPase–associated signaling cascades and affects Na/K-ATPase localization. The retinoschisin-Na/K-ATPase complex overlaps with signaling mediators. Defective Na/K-ATPase signaling by retinoschisin deficiency may promote retinal dystrophy. Mutations in the RS1 gene cause X-linked juvenile retinoschisis (XLRS), a hereditary retinal dystrophy. We recently showed that retinoschisin, the protein encoded by RS1, regulates ERK signaling and apoptosis in retinal cells. In this study, we explored an influence of retinoschisin on the functionality of the Na/K-ATPase, its interaction partner at retinal plasma membranes. We show that retinoschisin binding requires the β2-subunit of the Na/K-ATPase, whereas the α-subunit is exchangeable. Our investigations revealed no effect of retinoschisin on Na/K-ATPase–mediated ATP hydrolysis and ion transport. However, we identified an influence of retinoschisin on Na/K-ATPase–regulated signaling cascades and Na/K-ATPase localization. In addition to the known ERK deactivation, retinoschisin treatment of retinoschisin-deficient (Rs1h-/Y) murine retinal explants decreased activation of Src, an initial transmitter in Na/K-ATPase signal transduction, and of Ca2+ signaling marker Camk2. Immunohistochemistry on murine retinae revealed an overlap of the retinoschisin–Na/K-ATPase complex with proteins involved in Na/K-ATPase signaling, such as caveolin, phospholipase C, Src, and the IP3 receptor. Finally, retinoschisin treatment altered Na/K-ATPase localization in photoreceptors of Rs1h-/Y retinae. Taken together, our results suggest a regulatory effect of retinoschisin on Na/K-ATPase signaling and localization, whereas Na/K-ATPase-dysregulation caused by retinoschisin deficiency could represent an initial step in XLRS pathogenesis.
Collapse
Affiliation(s)
- Karolina Plössl
- Institute of Human Genetics, University of Regensburg, 93053 Regensburg, Germany
| | - Melanie Royer
- Institute of Human Genetics, University of Regensburg, 93053 Regensburg, Germany
| | - Sarah Bernklau
- Institute of Human Genetics, University of Regensburg, 93053 Regensburg, Germany
| | - Neslihan N Tavraz
- Institute of Chemistry, Technical University of Berlin, 10623 Berlin, Germany
| | - Thomas Friedrich
- Institute of Chemistry, Technical University of Berlin, 10623 Berlin, Germany
| | - Jens Wild
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, 93053 Regensburg, Germany
| | - Bernhard H F Weber
- Institute of Human Genetics, University of Regensburg, 93053 Regensburg, Germany
| | - Ulrike Friedrich
- Institute of Human Genetics, University of Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
27
|
Cui X, Xie Z. Protein Interaction and Na/K-ATPase-Mediated Signal Transduction. Molecules 2017; 22:molecules22060990. [PMID: 28613263 PMCID: PMC6152704 DOI: 10.3390/molecules22060990] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/01/2017] [Accepted: 06/02/2017] [Indexed: 02/05/2023] Open
Abstract
The Na/K-ATPase (NKA), or Na pump, is a member of the P-type ATPase superfamily. In addition to pumping ions across cell membrane, it is engaged in assembly of multiple protein complexes in the plasma membrane. This assembly allows NKA to perform many non-pumping functions including signal transduction that are important for animal physiology and disease progression. This article will focus on the role of protein interaction in NKA-mediated signal transduction, and its potential utility as target for developing new therapeutics.
Collapse
Affiliation(s)
- Xiaoyu Cui
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA.
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA.
| |
Collapse
|
28
|
Miao Y, Bhushan J, Dani A, Vig M. Na + influx via Orai1 inhibits intracellular ATP-induced mTORC2 signaling to disrupt CD4 T cell gene expression and differentiation. eLife 2017; 6. [PMID: 28492364 PMCID: PMC5459575 DOI: 10.7554/elife.25155] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/10/2017] [Indexed: 12/16/2022] Open
Abstract
T cell effector functions require sustained calcium influx. However, the signaling and phenotypic consequences of non-specific sodium permeation via calcium channels remain unknown. α-SNAP is a crucial component of Orai1 channels, and its depletion disrupts the functional assembly of Orai1 multimers. Here we show that α-SNAP hypomorph, hydrocephalus with hopping gait, Napahyh/hyh mice harbor significant defects in CD4 T cell gene expression and Foxp3 regulatory T cell (Treg) differentiation. Mechanistically, TCR stimulation induced rapid sodium influx in Napahyh/hyh CD4 T cells, which reduced intracellular ATP, [ATP]i. Depletion of [ATP]i inhibited mTORC2 dependent NFκB activation in Napahyh/hyh cells but ablation of Orai1 restored it. Remarkably, TCR stimulation in the presence of monensin phenocopied the defects in Napahyh/hyh signaling and Treg differentiation, but not IL-2 expression. Thus, non-specific sodium influx via bonafide calcium channels disrupts unexpected signaling nodes and may provide mechanistic insights into some divergent phenotypes associated with Orai1 function. DOI:http://dx.doi.org/10.7554/eLife.25155.001
Collapse
Affiliation(s)
- Yong Miao
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, United States
| | - Jaya Bhushan
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, United States
| | - Adish Dani
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, United States.,Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, United States
| | - Monika Vig
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, United States
| |
Collapse
|
29
|
Valvassori SS, Dal-Pont GC, Resende WR, Jornada LK, Peterle BR, Machado AG, Farias HR, de Souza CT, Carvalho AF, Quevedo J. Lithium and valproate act on the GSK-3β signaling pathway to reverse manic-like behavior in an animal model of mania induced by ouabain. Neuropharmacology 2017; 117:447-459. [DOI: 10.1016/j.neuropharm.2016.10.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 10/04/2016] [Accepted: 10/13/2016] [Indexed: 11/28/2022]
|
30
|
Na/K Pump and Beyond: Na/K-ATPase as a Modulator of Apoptosis and Autophagy. Molecules 2017; 22:molecules22040578. [PMID: 28430151 PMCID: PMC6154632 DOI: 10.3390/molecules22040578] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/29/2017] [Accepted: 03/29/2017] [Indexed: 01/22/2023] Open
Abstract
Lung cancer is a leading cause of global cancer deaths. Na/K-ATPase has been studied as a target for cancer treatment. Cardiotonic steroids (CS) trigger intracellular signalling upon binding to Na/K-ATPase. Normal lung and tumour cells frequently express different pump isoforms. Thus, Na/K-ATPase is a powerful target for lung cancer treatment. Drugs targeting Na/K-ATPase may induce apoptosis and autophagy in transformed cells. We argue that Na/K-ATPase has a role as a potential target in chemotherapy in lung cancer treatment. We discuss the effects of Na/K-ATPase ligands and molecular pathways inducing deleterious effects on lung cancer cells, especially those leading to apoptosis and autophagy.
Collapse
|
31
|
Hangaard L, Bouzinova EV, Staehr C, Dam VS, Kim S, Xie Z, Aalkjaer C, Matchkov VV. Na-K-ATPase regulates intercellular communication in the vascular wall via cSrc kinase-dependent connexin43 phosphorylation. Am J Physiol Cell Physiol 2017; 312:C385-C397. [DOI: 10.1152/ajpcell.00347.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/10/2017] [Accepted: 01/14/2017] [Indexed: 12/23/2022]
Abstract
Communication between vascular smooth muscle cells (VSMCs) is dependent on gap junctions and is regulated by the Na-K-ATPase. The Na-K-ATPase is therefore important for synchronized VSMC oscillatory activity, i.e., vasomotion. The signaling between the Na-K-ATPase and gap junctions is unknown. We tested here the hypothesis that this signaling involves cSrc kinase. Intercellular communication was assessed by membrane capacitance measurements of electrically coupled VSMCs. Vasomotion in isometric myograph, input resistance, and synchronized [Ca2+]i transients were used as readout for intercellular coupling in rat mesenteric small arteries in vitro. Phosphorylation of cSrc kinase and connexin43 (Cx43) were semiquantified by Western blotting. Micromole concentration of ouabain reduced the amplitude of norepinephrine-induced vasomotion and desynchronized Ca2+ transients in VSMC in the arterial wall. Ouabain also increased input resistance in the arterial wall. These effects of ouabain were antagonized by inhibition of tyrosine phosphorylation with genistein, PP2, and by an inhibitor of the Na-K-ATPase-dependent cSrc activation, pNaKtide. Moreover, inhibition of cSrc phosphorylation increased vasomotion amplitude and decreased the resistance between cells in the vascular wall. Ouabain inhibited the electrical coupling between A7r5 cells, but pNaKtide restored the electrical coupling. Ouabain increased cSrc autophosphorylation of tyrosine 418 (Y418) required for full catalytic activity whereas pNaKtide antagonized it. This cSrc activation was associated with Cx43 phosphorylation of tyrosine 265 (Y265). Our findings demonstrate that Na-K-ATPase regulates intercellular communication in the vascular wall via cSrc-dependent Cx43 tyrosine phosphorylation.
Collapse
Affiliation(s)
- Lise Hangaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | | - Vibeke S. Dam
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Sukhan Kim
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Christian Aalkjaer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, University of Copenhagen, Copenhagen, Denmark; and
| | | |
Collapse
|
32
|
Madan N, Xu Y, Duan Q, Banerjee M, Larre I, Pierre SV, Xie Z. Src-independent ERK signaling through the rat α3 isoform of Na/K-ATPase. Am J Physiol Cell Physiol 2017; 312:C222-C232. [PMID: 27903584 PMCID: PMC5401946 DOI: 10.1152/ajpcell.00199.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 11/30/2016] [Accepted: 11/30/2016] [Indexed: 01/10/2023]
Abstract
The Na/K-ATPase α1 polypeptide supports both ion-pumping and signaling functions. The Na/K-ATPase α3 polypeptide differs from α1 in both its primary structure and its tissue distribution. The expression of α3 seems particularly important in neurons, and recent clinical evidence supports a unique role of this isoform in normal brain function. The nature of this specific role of α3 has remained elusive, because the ubiquitous presence of α1 has hindered efforts to characterize α3-specific functions in mammalian cell systems. Using Na/K-ATPase α1 knockdown pig kidney cells (PY-17), we generated the first stable mammalian cell line expressing a ouabain-resistant form of rat Na/K-ATPase α3 in the absence of endogenous pig α1 detectable by Western blotting. In these cells, Na/K-ATPase α3 formed a functional ion-pumping enzyme and rescued the expression of Na/K-ATPase β1 and caveolin-1 to levels comparable with those observed in PY-17 cells rescued with a rat Na/K-ATPase α1 (AAC-19). The α3-containing enzymes had lower Na+ affinity and lower ouabain-sensitive transport activity than their α1-containing counterparts under basal conditions, but showed a greater capacity to be activated when intracellular Na+ was increased. In contrast to Na/K-ATPase α1, α3 could not regulate Src. Upon exposure to ouabain, Src activation did not occur, yet ERK was activated through Src-independent pathways involving PI3K and PKC. Hence, α3 expression confers signaling and pumping properties that are clearly distinct from that of cells expressing Na/K-ATPase α1.
Collapse
Affiliation(s)
- Namrata Madan
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Yunhui Xu
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China; and
| | - Qiming Duan
- Gladstone Institute of Cardiovascular Disease, San Francisco, California
| | - Moumita Banerjee
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Isabel Larre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia;
| |
Collapse
|
33
|
Abstract
Since the beginning of investigations of the Na,K-ATPase, it has been well-known that Mg2+ is an essential cofactor for activation of enzymatic ATP hydrolysis without being transported through the cell membrane. Moreover, experimental evidence has been collected through the years that shows that Mg2+ ions have a regulatory effect on ion transport by interacting with the cytoplasmic side of the ion pump. Our experiments allowed us to reveal the underlying mechanism. Mg2+ is able to bind to a site outside the membrane domain of the protein's α subunit, close to the entrance of the access channel to the ion-binding sites, thus modifying the local concentration of the ions in the electrolyte, of which Na+, K+, and H+ are of physiological interest. The decrease in the concentration of these cations can be explained by electrostatic interaction and estimated by the Debye-Hückel theory. This effect provokes the observed apparent reduction of the binding affinity of the binding sites of the Na,K-ATPase in the presence of various Mg2+ concentrations. The presence of the bound Mg2+, however, does not affect the reaction kinetics of the transport function of the ion pump. Therefore, stopped-flow experiments could be performed to gain the first insight into the Na+ binding kinetics on the cytoplasmic side by Mg2+ concentration jump experiments.
Collapse
Affiliation(s)
- Hans-Jürgen Apell
- Department of Biology, University of Konstanz , 78464 Konstanz, Germany
| | - Tanja Hitzler
- Department of Biology, University of Konstanz , 78464 Konstanz, Germany
| | - Grischa Schreiber
- Department of Biology, University of Konstanz , 78464 Konstanz, Germany
| |
Collapse
|
34
|
Plössl K, Weber BHF, Friedrich U. The X-linked juvenile retinoschisis protein retinoschisin is a novel regulator of mitogen-activated protein kinase signalling and apoptosis in the retina. J Cell Mol Med 2016; 21:768-780. [PMID: 27995734 PMCID: PMC5345684 DOI: 10.1111/jcmm.13019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/26/2016] [Indexed: 02/01/2023] Open
Abstract
X-linked juvenile retinoschisis (XLRS) is a hereditary retinal dystrophy in young males, caused by mutations in the RS1 gene. The function of the encoded protein, termed retinoschisin, and the molecular mechanisms underlying XLRS pathogenesis are still unresolved, although a direct interaction partner of the secreted retinoschisin, the retinal Na/K-ATPase, was recently identified. Earlier gene expression studies in retinoschisin-deficient (Rs1h-/Y ) mice provided a first indication of pathological up-regulation of mitogen-activated protein (MAP) kinase signalling in disease pathogenesis. To further investigate the role for retinoschisin in MAP kinase regulation, we exposed Y-79 cells and murine Rs1h-/Y retinae to recombinant retinoschisin and the XLRS-associated mutant RS1-C59S. Although normal retinoschisin stably bound to retinal cells, RS1-C59S exhibited a strongly reduced binding affinity. Simultaneously, exposure to normal retinoschisin significantly reduced phosphorylation of C-RAF and MAP kinases ERK1/2 in Y-79 cells and murine Rs1h-/Y retinae. Expression of MAP kinase target genes C-FOS and EGR1 was also down-regulated in both model systems. Finally, retinoschisin treatment decreased pro-apoptotic BAX-2 transcript levels in Y-79 cells and Rs1h-/Y retinae. Upon retinoschisin treatment, these cells showed increased resistance against apoptosis, reflected by decreased caspase-3 activity (in Y-79 cells) and increased photoreceptor survival (in Rs1h-/Y retinal explants). RS1-C59S did not influence C-RAF or ERK1/2 activation, C-FOS or EGR1 expression, or apoptosis. Our data imply that retinoschisin is a novel regulator of MAP kinase signalling and exerts an anti-apoptotic effect on retinal cells. We therefore discuss that disturbances of MAP kinase signalling by retinoschisin deficiency could be an initial step in XLRS pathogenesis.
Collapse
Affiliation(s)
- Karolina Plössl
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
| | - Bernhard H F Weber
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
| | - Ulrike Friedrich
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
| |
Collapse
|
35
|
Yan Y, Shapiro AP, Mopidevi BR, Chaudhry MA, Maxwell K, Haller ST, Drummond CA, Kennedy DJ, Tian J, Malhotra D, Xie ZJ, Shapiro JI, Liu J. Protein Carbonylation of an Amino Acid Residue of the Na/K-ATPase α1 Subunit Determines Na/K-ATPase Signaling and Sodium Transport in Renal Proximal Tubular Cells. J Am Heart Assoc 2016; 5:e003675. [PMID: 27613772 PMCID: PMC5079028 DOI: 10.1161/jaha.116.003675] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 08/12/2016] [Indexed: 01/12/2023]
Abstract
BACKGROUND We have demonstrated that cardiotonic steroids, such as ouabain, signaling through the Na/K-ATPase, regulate sodium reabsorption in the renal proximal tubule. By direct carbonylation modification of the Pro222 residue in the actuator (A) domain of pig Na/K-ATPase α1 subunit, reactive oxygen species are required for ouabain-stimulated Na/K-ATPase/c-Src signaling and subsequent regulation of active transepithelial (22)Na(+) transport. In the present study we sought to determine the functional role of Pro222 carbonylation in Na/K-ATPase signaling and sodium handling. METHODS AND RESULTS Stable pig α1 knockdown LLC-PK1-originated PY-17 cells were rescued by expressing wild-type rat α1 and rat α1 with a single mutation of Pro224 (corresponding to pig Pro222) to Ala. This mutation does not affect ouabain-induced inhibition of Na/K-ATPase activity, but abolishes the effects of ouabain on Na/K-ATPase/c-Src signaling, protein carbonylation, Na/K-ATPase endocytosis, and active transepithelial (22)Na(+) transport. CONCLUSIONS Direct carbonylation modification of Pro224 in the rat α1 subunit determines ouabain-mediated Na/K-ATPase signal transduction and subsequent regulation of renal proximal tubule sodium transport.
Collapse
Affiliation(s)
- Yanling Yan
- Department of Pharmacology, Physiology and Toxicology, JCE School of Medicine, Marshall University, Huntington, WV
| | - Anna P Shapiro
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH
| | - Brahma R Mopidevi
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH
| | - Muhammad A Chaudhry
- Department of Pharmacology, Physiology and Toxicology, JCE School of Medicine, Marshall University, Huntington, WV
| | - Kyle Maxwell
- Department of Pharmacology, Physiology and Toxicology, JCE School of Medicine, Marshall University, Huntington, WV
| | - Steven T Haller
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH
| | | | - David J Kennedy
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH
| | - Jiang Tian
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH
| | - Deepak Malhotra
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH
| | - Zi-Jian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Joseph I Shapiro
- Department of Pharmacology, Physiology and Toxicology, JCE School of Medicine, Marshall University, Huntington, WV Department of Medicine, University of Toledo College of Medicine, Toledo, OH
| | - Jiang Liu
- Department of Pharmacology, Physiology and Toxicology, JCE School of Medicine, Marshall University, Huntington, WV Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| |
Collapse
|
36
|
Liu L, Wu J, Kennedy DJ. Regulation of Cardiac Remodeling by Cardiac Na(+)/K(+)-ATPase Isoforms. Front Physiol 2016; 7:382. [PMID: 27667975 PMCID: PMC5016610 DOI: 10.3389/fphys.2016.00382] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/22/2016] [Indexed: 12/20/2022] Open
Abstract
Cardiac remodeling occurs after cardiac pressure/volume overload or myocardial injury during the development of heart failure and is a determinant of heart failure. Preventing or reversing remodeling is a goal of heart failure therapy. Human cardiomyocyte Na+/K+-ATPase has multiple α isoforms (1–3). The expression of the α subunit of the Na+/K+-ATPase is often altered in hypertrophic and failing hearts. The mechanisms are unclear. There are limited data from human cardiomyocytes. Abundant evidences from rodents show that Na+/K+-ATPase regulates cardiac contractility, cell signaling, hypertrophy and fibrosis. The α1 isoform of the Na+/K+-ATPase is the ubiquitous isoform and possesses both pumping and signaling functions. The α2 isoform of the Na+/K+-ATPase regulates intracellular Ca2+ signaling, contractility and pathological hypertrophy. The α3 isoform of the Na+/K+-ATPase may also be a target for cardiac hypertrophy. Restoration of cardiac Na+/K+-ATPase expression may be an effective approach for prevention of cardiac remodeling. In this article, we will overview: (1) the distribution and function of isoform specific Na+/K+-ATPase in the cardiomyocytes. (2) the role of cardiac Na+/K+-ATPase in the regulation of cell signaling, contractility, cardiac hypertrophy and fibrosis in vitro and in vivo. Selective targeting of cardiac Na+/K+-ATPase isoform may offer a new target for the prevention of cardiac remodeling.
Collapse
Affiliation(s)
- Lijun Liu
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo Toledo, OH, USA
| | - Jian Wu
- Center for Craniofacial Molecular Biology, University of Southern California Los Angeles, CA, USA
| | - David J Kennedy
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo Toledo, OH, USA
| |
Collapse
|
37
|
Srikanthan K, Shapiro JI, Sodhi K. The Role of Na/K-ATPase Signaling in Oxidative Stress Related to Obesity and Cardiovascular Disease. Molecules 2016; 21:molecules21091172. [PMID: 27598118 PMCID: PMC5642908 DOI: 10.3390/molecules21091172] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 08/26/2016] [Accepted: 09/01/2016] [Indexed: 12/16/2022] Open
Abstract
Na/K-ATPase has been extensively studied for its ion pumping function, but, in the past several decades, has been identified as a scaffolding and signaling protein. Initially it was found that cardiotonic steroids (CTS) mediate signal transduction through the Na/K-ATPase and result in the generation of reactive oxygen species (ROS), which are also capable of initiating the signal cascade. However, in recent years, this Na/K-ATPase/ROS amplification loop has demonstrated significance in oxidative stress related disease states, including obesity, atherosclerosis, heart failure, uremic cardiomyopathy, and hypertension. The discovery of this novel oxidative stress signaling pathway, holds significant therapeutic potential for the aforementioned conditions and others that are rooted in ROS.
Collapse
Affiliation(s)
- Krithika Srikanthan
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA.
| | - Joseph I Shapiro
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA.
| | - Komal Sodhi
- Department of Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA.
| |
Collapse
|
38
|
Matchkov VV, Krivoi II. Specialized Functional Diversity and Interactions of the Na,K-ATPase. Front Physiol 2016; 7:179. [PMID: 27252653 PMCID: PMC4879863 DOI: 10.3389/fphys.2016.00179] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/04/2016] [Indexed: 12/22/2022] Open
Abstract
Na,K-ATPase is a protein ubiquitously expressed in the plasma membrane of all animal cells and vitally essential for their functions. A specialized functional diversity of the Na,K-ATPase isozymes is provided by molecular heterogeneity, distinct subcellular localizations, and functional interactions with molecular environment. Studies over the last decades clearly demonstrated complex and isoform-specific reciprocal functional interactions between the Na,K-ATPase and neighboring proteins and lipids. These interactions are enabled by a spatially restricted ion homeostasis, direct protein-protein/lipid interactions, and protein kinase signaling pathways. In addition to its "classical" function in ion translocation, the Na,K-ATPase is now considered as one of the most important signaling molecules in neuronal, epithelial, skeletal, cardiac and vascular tissues. Accordingly, the Na,K-ATPase forms specialized sub-cellular multimolecular microdomains which act as receptors to circulating endogenous cardiotonic steroids (CTS) triggering a number of signaling pathways. Changes in these endogenous cardiotonic steroid levels and initiated signaling responses have significant adaptive values for tissues and whole organisms under numerous physiological and pathophysiological conditions. This review discusses recent progress in the studies of functional interactions between the Na,K-ATPase and molecular microenvironment, the Na,K-ATPase-dependent signaling pathways and their significance for diversity of cell function.
Collapse
Affiliation(s)
| | - Igor I Krivoi
- Department of General Physiology, St. Petersburg State University St. Petersburg, Russia
| |
Collapse
|
39
|
Yosef E, Katz A, Peleg Y, Mehlman T, Karlish SJD. Do Src Kinase and Caveolin Interact Directly with Na,K-ATPase? J Biol Chem 2016; 291:11736-50. [PMID: 27022017 DOI: 10.1074/jbc.m116.721084] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Indexed: 12/14/2022] Open
Abstract
Much evidence points to a role of Na,K-ATPase in ouabain-dependent signal transduction. Based on experiments with different cell lines and native tissue membranes, a current hypothesis postulates direct interactions between the Na,K-ATPase and Src kinase (non-receptor tyrosine kinase). Na,K-ATPase is proposed to bind Src kinase and inhibit its activity, whereas ouabain, the specific Na,K-ATPase inhibitor, binds and stabilizes the E2 conformation, thus exposing the Src kinase domain and its active site Tyr-418 for activation. Ouabain-dependent signaling is thought to be mediated within caveolae by a complex consisting of Na,K-ATPase, caveolin, and Src kinase. In the current work, we have looked for direct interactions utilizing purified recombinant Na,K-ATPase (human α1β1FXYD1 or porcine α1D369Nβ1FXYD1) and purified human Src kinase and human caveolin 1 or interactions between these proteins in native membrane vesicles isolated from rabbit kidney. By several independent criteria and techniques, no stable interactions were detected between Na,K-ATPase and purified Src kinase. Na,K-ATPase was found to be a substrate for Src kinase phosphorylation at Tyr-144. Clear evidence for a direct interaction between purified human Na,K-ATPase and human caveolin was obtained, albeit with a low molar stoichiometry (1:15-30 caveolin 1/Na,K-ATPase). In native renal membranes, a specific caveolin 14-5 oligomer (95 kDa) was found to be in direct interaction with Na,K-ATPase. We inferred that a small fraction of the renal Na,K-ATPase molecules is in a ∼1:1 complex with a caveolin 14-5 oligomer. Thus, overall, whereas a direct caveolin 1/Na,K-ATPase interaction is confirmed, the lack of direct Src kinase/Na,K-ATPase binding requires reassessment of the mechanism of ouabain-dependent signaling.
Collapse
Affiliation(s)
| | | | - Yoav Peleg
- the Israel Structural Proteomics Center, and
| | - Tevie Mehlman
- the Biological Services Department-Mass Spectrometry unit, Weizmann Institute of Science, Rehovot 7610001, Israel
| | | |
Collapse
|
40
|
Morrill GA, Kostellow AB, Liu L, Gupta RK, Askari A. Evolution of the α-Subunit of Na/K-ATPase from Paramecium to Homo sapiens: Invariance of Transmembrane Helix Topology. J Mol Evol 2016; 82:183-98. [PMID: 26961431 PMCID: PMC4866997 DOI: 10.1007/s00239-016-9732-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/03/2016] [Indexed: 12/01/2022]
Abstract
Na/K-ATPase is a key plasma membrane enzyme involved in cell signaling, volume regulation, and maintenance of electrochemical gradients. The α-subunit, central to these functions, belongs to a large family of P-type ATPases. Differences in transmembrane (TM) helix topology, sequence homology, helix–helix contacts, cell signaling, and protein domains of Na/K-ATPase α-subunit were compared in fungi (Beauveria), unicellular organisms (Paramecia), primitive multicellular organisms (Hydra), and vertebrates (Xenopus, Homo sapiens), and correlated with evolution of physiological functions in the α-subunit. All α-subunits are of similar length, with groupings of four and six helices in the N- and C-terminal regions, respectively. Minimal homology was seen for protein domain patterns in Paramecium and Hydra, with high correlation between Hydra and vertebrates. Paramecium α-subunits display extensive disorder, with minimal helix contacts. Increases in helix contacts in Hydra approached vertebrates. Protein motifs known to be associated with membrane lipid rafts and cell signaling reveal significant positional shifts between Paramecium and Hydra vulgaris, indicating that regional membrane fluidity changes occur during evolution. Putative steroid binding sites overlapping TM-3 occurred in all species. Sites associated with G-protein-receptor stimulation occur both in vertebrates and amphibia but not in Hydra or Paramecia. The C-terminus moiety “KETYY,” necessary for the Na+ activation of pump phosphorylation, is not present in unicellular species indicating the absence of classical Na+/K+-pumps. The basic protein topology evolved earliest, followed by increases in protein domains and ordered helical arrays, correlated with appearance of α-subunit regions known to involve cell signaling, membrane recycling, and ion channel formation.
Collapse
Affiliation(s)
- Gene A Morrill
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| | - Adele B Kostellow
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Lijun Liu
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH, 43614, USA
| | - Raj K Gupta
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Amir Askari
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH, 43614, USA
| |
Collapse
|
41
|
Durlacher CT, Chow K, Chen XW, He ZX, Zhang X, Yang T, Zhou SF. Targeting Na⁺/K⁺ -translocating adenosine triphosphatase in cancer treatment. Clin Exp Pharmacol Physiol 2016; 42:427-43. [PMID: 25739707 DOI: 10.1111/1440-1681.12385] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 02/09/2015] [Accepted: 02/21/2015] [Indexed: 12/24/2022]
Abstract
The Na(+) /K(+) -translocating adenosine triphosphatase (ATPase) transports sodium and potassium across the plasma membrane and represents a potential target in cancer chemotherapy. Na(+) /K(+) -ATPase belongs to the P-type ATPase family (also known as E1-E2 ATPase), which is involved in transporting certain ions, metals, and lipids across the plasma membrane of mammalian cells. In humans, the Na(+) /K(+) -ATPase is a binary complex of an α-subunit that has four isoforms (α1 -α4 ) and a β-subunit that has three isoforms (β1 -β3 ). This review aims to update our knowledge on the role of Na(+) /K(+) -ATPase in cancer development and metastasis, as well as on how Na(+) /K(+) -ATPase inhibitors kill tumour cells. The Na(+) /K(+) -ATPase has been found to be associated with cancer initiation, growth, development, and metastasis. Cardiac glycosides have exhibited anticancer effects in cell-based and mouse studies via inhibition of the Na(+) /K(+) -ATPase and other mechanisms. Na(+) /K(+) -ATPase inhibitors may kill cancer cells via induction of apoptosis and autophagy, radical oxygen species production, and cell cycle arrest. They also modulate multiple signalling pathways that regulate cancer cell survival and death, which contributes to their antiproliferative activities in cancer cells. The clinical evidence supporting the use of Na(+) /K(+) -ATPase inhibitors as anticancer drugs is weak. Several phase I and phase II clinical trials with digoxin, Anvirzel, and huachansu (an intravenous formulated extract of the venom of the wild toad), either alone or more often in combination with other anticancer agents, have shown acceptable safety profiles but limited efficacy in cancer patients. Well-designed randomized clinical trials with reasonable sample sizes are certainly warranted to confirm the efficacy and safety of cardiac glycosides for the treatment of cancer.
Collapse
Affiliation(s)
- Cameron T Durlacher
- Department of Pharmaceutical Science, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Aperia A, Akkuratov EE, Fontana JM, Brismar H. Na+-K+-ATPase, a new class of plasma membrane receptors. Am J Physiol Cell Physiol 2016; 310:C491-5. [PMID: 26791490 DOI: 10.1152/ajpcell.00359.2015] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The Na(+)-K(+)-ATPase (NKA) differs from most other ion transporters, not only in its capacity to maintain a steep electrochemical gradient across the plasma membrane, but also as a receptor for a family of cardiotonic steroids, to which ouabain belongs. Studies from many groups, performed during the last 15 years, have demonstrated that ouabain, a member of the cardiotonic steroid family, can activate a network of signaling molecules, and that NKA will also serve as a signal transducer that can provide a feedback loop between NKA and the mitochondria. This brief review summarizes the current knowledge and controversies with regard to the understanding of NKA signaling.
Collapse
Affiliation(s)
- Anita Aperia
- Science for Life Laboratory, Department of Women and Children's Health, Karolinska Institutet, Stockholm, Sweden; and
| | - Evgeny E Akkuratov
- Science for Life Laboratory, Department of Women and Children's Health, Karolinska Institutet, Stockholm, Sweden; and
| | - Jacopo Maria Fontana
- Science for Life Laboratory, Department of Applied Physics, Royal Institute of Technology, Stockholm, Sweden
| | - Hjalmar Brismar
- Science for Life Laboratory, Department of Women and Children's Health, Karolinska Institutet, Stockholm, Sweden; and Science for Life Laboratory, Department of Applied Physics, Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
43
|
Siriwardana G, Seligman PA. Iron depletion results in Src kinase inhibition with associated cell cycle arrest in neuroblastoma cells. Physiol Rep 2015; 3:3/3/e12341. [PMID: 25825542 PMCID: PMC4393172 DOI: 10.14814/phy2.12341] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Iron is required for cellular proliferation. Recently, using systematic time studies of neuroblastoma cell growth, we better defined the G1 arrest caused by iron chelation to a point in mid-G1, where cyclin E protein is present, but the cyclin E/CDK2 complex kinase activity is inhibited. In this study, we again used the neuroblastoma SKNSH cells lines to pinpoint the mechanism responsible for this G1 block. Initial studies showed in the presence of DFO, these cells have high levels of p27 and after reversal of iron chelation p27 is degraded allowing for CDK2 kinase activity. The initial activation of CDK2 kinase allows cells to exit G1 and enter S phase. Furthermore, we found that inhibition of p27 degradation by DFO is directly associated with inhibition of Src kinase activity measured by lack of phosphorylation of Src at the 416 residue. Activation of Src kinase occurs very early after reversal from the DFO G1 block and is temporally associated with initiation of cellular proliferation associated with entry into S phase. For the first time therefore we show that iron chelation inhibits Src kinase activity and this activity is a requirement for cellular proliferation.
Collapse
Affiliation(s)
- Gamini Siriwardana
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado
| | - Paul A Seligman
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
44
|
Wang CW, Huang YC, Chan FN, Su SC, Kuo YH, Huang SF, Hung MW, Lin HC, Chang WL, Chang TC. A gut microbial metabolite of ginsenosides, compound K, induces intestinal glucose absorption and Na(+) /glucose cotransporter 1 gene expression through activation of cAMP response element binding protein. Mol Nutr Food Res 2015; 59:670-84. [PMID: 25600494 DOI: 10.1002/mnfr.201400688] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 12/13/2014] [Accepted: 12/30/2014] [Indexed: 01/21/2023]
Abstract
SCOPE The Na(+) /glucose cotransporter 1 (SGLT1) plays a crucial role in glucose uptake in intestinal epithelial cells (IECs), which has been shown essential in ameliorating intestinal inflammation. Ginseng has historically been used to treat inflammatory disorders. Understanding the regulatory mechanism of ginseng-mediated induction of SGLT1 gene expression in human intestinal cells is therefore important. METHODS AND RESULTS We demonstrate that ginsenoside compound K (CK) enhances SGLT1-mediated glucose uptake in mice and human intestinal Caco-2 cells. Transient transfection analysis using SGLT1 promoter-luciferase reporters demonstrated that the presence of an essential cAMP response element (CRE) is required for CK-mediated induction of SGLT1 gene expression. The ChIP assays indicated that increased CRE-binding protein (CREB) and CREB-binding protein (CBP) binding to the SGLT1 promoter in CK-treated cells is associated with an activated chromatin state. Our result showed that the increased CREB phosphorylation is directly correlated with SGLT1 expression in IECs. Further studies indicated that the epidermal growth factor receptor (EGFR) signaling pathway is involved in the CK-mediated effect. CONCLUSION These findings provide a novel mechanism for the CK-mediated upregulation of SGLT1 expression through EGFR-CREB signaling activation, which could contribute to reducing gut inflammation.
Collapse
Affiliation(s)
- Chun-Wen Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Lauf PK, Alqahtani T, Flues K, Meller J, Adragna NC. Interaction between Na-K-ATPase and Bcl-2 proteins BclXL and Bak. Am J Physiol Cell Physiol 2015; 308:C51-60. [DOI: 10.1152/ajpcell.00287.2014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In silico analysis predicts interaction between Na-K-ATPase (NKA) and Bcl-2 protein canonical BH3- and BH1-like motifs, consistent with NKA inhibition by the benzo-phenanthridine alkaloid chelerythrine, a BH3 mimetic, in fetal human lens epithelial cells (FHLCs) (Lauf PK, Heiny J, Meller J, Lepera MA, Koikov L, Alter GM, Brown TL, Adragna NC. Cell Physiol Biochem 31: 257–276, 2013). This report establishes proof of concept: coimmunoprecipitation and immunocolocalization showed unequivocal and direct physical interaction between NKA and Bcl-2 proteins. Specifically, NKA antibodies (ABs) coimmunoprecipitated BclXL (B-cell lymphoma extra large) and BAK (Bcl-2 antagonist killer) proteins in FHLCs and A549 lung cancer cells. In contrast, both anti-Bcl-2 ABs failed to pull down NKA. Notably, the molecular mass of BAK1 proteins pulled down by NKA and BclXL ABs appeared to be some 4-kDa larger than found in input monomers. In silico analysis predicts these higher molecular mass BAK1 proteins as alternative splicing variants, encoding 42 amino acid (aa) larger proteins than the known 211-aa long canonical BAK1 protein. These BAK1 variants may constitute a pool separate from that forming mitochondrial pores by specifically interacting with NKA and BclXL proteins. We propose a NKA-Bcl-2 protein ternary complex supporting our hypothesis for a special sensor role of NKA in Bcl-2 protein control of cell survival and apoptosis.
Collapse
Affiliation(s)
- Peter K. Lauf
- Cell Biophysics Group, Wright State University Boonshoft School of Medicine, Dayton, Ohio
- Department of Pathology, Wright State University Boonshoft School of Medicine, Dayton, Ohio
- Department of Pharmacology and Toxicology, Wright State University Boonshoft School of Medicine, Dayton, Ohio; and
| | - Tariq Alqahtani
- Cell Biophysics Group, Wright State University Boonshoft School of Medicine, Dayton, Ohio
- Department of Pharmacology and Toxicology, Wright State University Boonshoft School of Medicine, Dayton, Ohio; and
| | - Karin Flues
- Cell Biophysics Group, Wright State University Boonshoft School of Medicine, Dayton, Ohio
- Department of Pharmacology and Toxicology, Wright State University Boonshoft School of Medicine, Dayton, Ohio; and
| | - Jaroslaw Meller
- Environmental Health, University of Cincinnati Medical Center, Cincinnati, Ohio
| | - Norma C. Adragna
- Cell Biophysics Group, Wright State University Boonshoft School of Medicine, Dayton, Ohio
- Department of Pharmacology and Toxicology, Wright State University Boonshoft School of Medicine, Dayton, Ohio; and
| |
Collapse
|
46
|
Xie M, Chen D, Zhang F, Willsky GR, Crans DC, Ding W. Effects of vanadium (III, IV, V)-chlorodipicolinate on glycolysis and antioxidant status in the liver of STZ-induced diabetic rats. J Inorg Biochem 2014; 136:47-56. [DOI: 10.1016/j.jinorgbio.2014.03.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Revised: 03/23/2014] [Accepted: 03/23/2014] [Indexed: 02/01/2023]
|
47
|
Clifford RJ, Kaplan JH. Human breast tumor cells are more resistant to cardiac glycoside toxicity than non-tumorigenic breast cells. PLoS One 2013; 8:e84306. [PMID: 24349570 PMCID: PMC3862803 DOI: 10.1371/journal.pone.0084306] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 11/13/2013] [Indexed: 12/28/2022] Open
Abstract
Cardiotonic steroids (CTS), specific inhibitors of Na,K-ATPase activity, have been widely used for treating cardiac insufficiency. Recent studies suggest that low levels of endogenous CTS do not inhibit Na,K-ATPase activity but play a role in regulating blood pressure, inducing cellular kinase activity, and promoting cell viability. Higher CTS concentrations inhibit Na,K-ATPase activity and can induce reactive oxygen species, growth arrest, and cell death. CTS are being considered as potential novel therapies in cancer treatment, as they have been shown to limit tumor cell growth. However, there is a lack of information on the relative toxicity of tumor cells and comparable non-tumor cells. We have investigated the effects of CTS compounds, ouabain, digitoxin, and bufalin, on cell growth and survival in cell lines exhibiting the full spectrum of non-cancerous to malignant phenotypes. We show that CTS inhibit membrane Na,K-ATPase activity equally well in all cell lines tested regardless of metastatic potential. In contrast, the cellular responses to the drugs are different in non-tumor and tumor cells. Ouabain causes greater inhibition of proliferation and more extensive apoptosis in non-tumor breast cells compared to malignant or oncogene-transfected cells. In tumor cells, the effects of ouabain are accompanied by activation of anti-apoptotic ERK1/2. However, ERK1/2 or Src inhibition does not sensitize tumor cells to CTS cytotoxicity, suggesting that other mechanisms provide protection to the tumor cells. Reduced CTS-sensitivity in breast tumor cells compared to non-tumor cells indicates that CTS are not good candidates as cancer therapies.
Collapse
Affiliation(s)
- Rebecca J. Clifford
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Jack H. Kaplan
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
48
|
Xie JX, Li X, Xie Z. Regulation of renal function and structure by the signaling Na/K-ATPase. IUBMB Life 2013; 65:991-8. [PMID: 24323927 PMCID: PMC5375025 DOI: 10.1002/iub.1229] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 11/08/2013] [Indexed: 12/23/2022]
Abstract
The Na/K-ATPase as an essential ion pump was discovered more than 50 years ago (Skou (1989) Biochim. Biophys. Acta 1000, 439-446; Feraille and Doucet (2001) Physiol. Rev. 81, 345-418). The signaling function of Na/K-ATPase has been gradually appreciated over the last 20 years, first from the studies of regulatory effects of ouabain on cardiac cell growth. Several reviews on this topic have been written during the last few years (Schoner and Scheiner-Bobis (2007) Am. J. Physiol. Cell. Physiol. 293, C509-C536; Xie and Cai (2003) Mol. Interv. 3, 157 - 168; Bagrov et al. (2009) Pharmacol. Rev. 61, 9-38; Tian and Xie (2008) Physiology 23, 205-211; Fontana et al. (2013) FEBS J. 280, 5450-5455; Blanco and Wallace (2013) Am. J. Physiol. Renal Physiol. 305, F797-F812). This article will focus on the molecular mechanism of Na/K-ATPase-mediated signal transduction and its potential regulatory role in renal physiology and diseases.
Collapse
Affiliation(s)
- Jeffrey X Xie
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, USA
| | | | | |
Collapse
|
49
|
Wu J, Akkuratov EE, Bai Y, Gaskill CM, Askari A, Liu L. Cell signaling associated with Na(+)/K(+)-ATPase: activation of phosphatidylinositide 3-kinase IA/Akt by ouabain is independent of Src. Biochemistry 2013; 52:9059-67. [PMID: 24266852 PMCID: PMC3868411 DOI: 10.1021/bi4011804] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Exposure
of intact cells to selective inhibitors of Na+/K+-ATPase such as ouabain activates several growth-related
cell signaling pathways. It has been suggested that the initial event
of these pathways is the binding of ouabain to a preexisting complex
of Src with Na+/K+-ATPase of the plasma membrane.
The aim of this work was to evaluate the role of Src in the ouabain-induced
activation of phosphatidylinositide 3-kinase 1A (PI3K1A) and its downstream
consequences. When fibroblasts devoid of Src (SYF cells) and controls
(Src++ cells) were exposed to ouabain, PI3K1A, Akt, and
proliferative growth were similarly stimulated in both cell lines.
Ouabain-induced activation of Akt was not prevented by the Src inhibitor
PP2. In contrast, ERK1/2 were not activated by ouabain in SYF cells
but were stimulated in Src++ cells; this was prevented
by PP2. In isolated adult mouse cardiac myocytes, where ouabain induces
hypertrophic growth, PP2 also did not prevent ouabain-induced activation
of Akt and the resulting hypertrophy. Ouabain-induced increases in
the levels of co-immunoprecipitation of the α-subunit of Na+/K+-ATPase with the p85 subunit of PI3K1A were
noted in SYF cells, Src++ cells, and adult cardiac myocytes.
In conjunction with previous findings, the results presented here
indicate that (a) if there is a preformed complex of Src and Na+/K+-ATPase, it is irrelevant to ouabain-induced
activation of the PI3K1A/Akt pathway through Na+/K+-ATPase and (b) a more likely, but not established, mechanism
of linkage of Na+/K+-ATPase to PI3K1A is the
ouabain-induced interaction of a proline-rich domain of the α-subunit
of Na+/K+-ATPase with the SH3 domain of the
p85 subunit of PI3K1A.
Collapse
Affiliation(s)
- Jian Wu
- Department of Biochemistry and Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus , Toledo, Ohio 43614, United States
| | | | | | | | | | | |
Collapse
|
50
|
Reinhard L, Tidow H, Clausen MJ, Nissen P. Na(+),K (+)-ATPase as a docking station: protein-protein complexes of the Na(+),K (+)-ATPase. Cell Mol Life Sci 2013; 70:205-22. [PMID: 22695678 PMCID: PMC11113973 DOI: 10.1007/s00018-012-1039-9] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Revised: 05/13/2012] [Accepted: 05/23/2012] [Indexed: 12/13/2022]
Abstract
The Na(+),K(+)-ATPase, or sodium pump, is well known for its role in ion transport across the plasma membrane of animal cells. It carries out the transport of Na(+) ions out of the cell and of K(+) ions into the cell and thus maintains electrolyte and fluid balance. In addition to the fundamental ion-pumping function of the Na(+),K(+)-ATPase, recent work has suggested additional roles for Na(+),K(+)-ATPase in signal transduction and biomembrane structure. Several signaling pathways have been found to involve Na(+),K(+)-ATPase, which serves as a docking station for a fast-growing number of protein interaction partners. In this review, we focus on Na(+),K(+)-ATPase as a signal transducer, but also briefly discuss other Na(+),K(+)-ATPase protein-protein interactions, providing a comprehensive overview of the diverse signaling functions ascribed to this well-known enzyme.
Collapse
Affiliation(s)
- Linda Reinhard
- Danish National Research Foundation, Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Department of Molecular Biology and Genetics, 8000 Aarhus C, Denmark
| | - Henning Tidow
- Danish National Research Foundation, Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Department of Molecular Biology and Genetics, 8000 Aarhus C, Denmark
| | - Michael J. Clausen
- Danish National Research Foundation, Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Department of Molecular Biology and Genetics, 8000 Aarhus C, Denmark
| | - Poul Nissen
- Danish National Research Foundation, Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Department of Molecular Biology and Genetics, 8000 Aarhus C, Denmark
| |
Collapse
|