1
|
Poluektov YM, Lopina OD, Strelkova MA, Kuleshova ID, Makarov AA, Petrushanko IY. Mechanisms mediating effects of cardiotonic steroids in mammalian blood cells. Front Pharmacol 2025; 16:1520927. [PMID: 40196366 PMCID: PMC11973394 DOI: 10.3389/fphar.2025.1520927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/21/2025] [Indexed: 04/09/2025] Open
Abstract
Cardiotonic steroids (CTSs) were known as steroidal plant compounds that exert cellular effects by the binding to Na,K-ATPase. Earlier, plant (exogenous) CTSs were used to treat chronic heart failure. By now, endogenous CTS have been identified in mammals, and their concentrations in the blood, normally in a subnanomolar range, are altered in numerous pathologies. This indicates their role as endogenous regulators of physiological processes. CTS transport occurs primarily in the blood, yet the CTS effects on blood cells remain poorly understood. This review summarizes the CTS effects on blood cells of animals and humans under normal and pathological conditions, and analyzes their action based on known mechanisms of action in mammalian cells. At high concentrations (greater than 10-9 M), CTS binding to Na,K-ATPase inhibits the enzyme, whereas lower concentrations of CTSs induce signaling cascades or activate the enzyme. All these mechanisms are shown to be present in blood cells. The particular CTS effect is determined by the CTS type, its concentration, the isoform composition of the catalytic α-subunit of Na,K-ATPase in the cell, and other cell features. It has been demonstrated that all blood cell types (erythrocytes, leukocytes, and platelets) expressed both ubiquitously distributed α1-isoform and tissue-specific α3-subunit, which exhibits a different ion and CTS affinity compared to α1. This results in a wide spectrum of blood cell responses to fluctuations in CTS levels in the blood. In particular, an increase in the level of endogenous CTSs by a more twofold is sufficient to induce a decline in the activity of erythrocyte Na,K-ATPase. The administration of exogenous CTSs is able to modulate the proinflammatory activity of leukocytes, which is attributed to the activation of signaling cascades, and to exert an influence on platelet activation. Hence, alterations of CTS levels in bloodstream significantly affect the functionality of blood cells, contributing to the organism's adaptive response. On top of this, a comparison of the effects of CTSs on human leukocytes and rodent leukocytes carrying the CTS-resistant α1-isoform often reveals opposite effects, thus indicating that rodents are an unsuitable model for studying CTS effects on these cells.
Collapse
Affiliation(s)
- Yuri M. Poluektov
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Olga D. Lopina
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Maria A. Strelkova
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Iuliia D. Kuleshova
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Alexander A. Makarov
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Irina Yu. Petrushanko
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
2
|
Whiteman NK. Evolution in small steps and giant leaps. Evolution 2022; 76:67-77. [PMID: 35040122 PMCID: PMC9387839 DOI: 10.1111/evo.14432] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/28/2021] [Accepted: 01/03/2022] [Indexed: 02/03/2023]
Abstract
The first Editor of Evolution was Ernst Mayr. His foreword to the first issue of Evolution published in 1947 framed evolution as a "problem of interaction" that was just beginning to be studied in this broad context. First, I explore progress and prospects on understanding the subsidiary interactions identified by Mayr, including interactions between parts of organisms, between individuals and populations, between species, and between the organism and its abiotic environment. Mayr's overall "problem of interaction" framework is examined in the context of coevolution within and among levels of biological organization. This leads to a comparison in the relative roles of biotic versus abiotic agents of selection and fluctuating versus directional selection, followed by stabilizing selection in shaping the genomic architecture of adaptation. Oligogenic architectures may be typical for traits shaped more by fluctuating selection and biotic selection. Conversely, polygenic architectures may be typical for traits shaped more by directional followed by stabilizing selection and abiotic selection. The distribution of effect sizes and turnover dynamics of adaptive alleles in these scenarios deserves further study. Second, I review two case studies on the evolution of acquired toxicity in animals, one involving cardiac glycosides obtained from plants and one involving bacterial virulence factors horizontally transferred to animals. The approaches used in these studies and the results gained directly flow from Mayr's vision of an evolutionary biology that revolves around the "problem of interaction."
Collapse
Affiliation(s)
- Noah K. Whiteman
- Department of Integrative Biology, University of California, Berkeley, California 94720
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720
| |
Collapse
|
3
|
Fedosova NU, Habeck M, Nissen P. Structure and Function of Na,K-ATPase-The Sodium-Potassium Pump. Compr Physiol 2021; 12:2659-2679. [PMID: 34964112 DOI: 10.1002/cphy.c200018] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Na,K-ATPase is an ubiquitous enzyme actively transporting Na-ions out of the cell in exchange for K-ions, thereby maintaining their concentration gradients across the cell membrane. Since its discovery more than six decades ago the Na-pump has been studied extensively and its vital physiological role in essentially every cell has been established. This article aims at providing an overview of well-established biochemical properties with a focus on Na,K-ATPase isoforms, its transport mechanism and principle conformations, inhibitors, and insights gained from crystal structures. © 2021 American Physiological Society. Compr Physiol 11:1-21, 2021.
Collapse
Affiliation(s)
| | - Michael Habeck
- Department of Molecular Biology and Genetics, Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Poul Nissen
- Department of Molecular Biology and Genetics, Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
4
|
Role of Na +/K +-ATPase in ischemic stroke: in-depth perspectives from physiology to pharmacology. J Mol Med (Berl) 2021; 100:395-410. [PMID: 34839371 DOI: 10.1007/s00109-021-02143-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 08/27/2021] [Accepted: 09/20/2021] [Indexed: 12/26/2022]
Abstract
Na+/K+-ATPase (NKA) is a large transmembrane protein expressed in all cells. It is well studied for its ion exchanging function, which is indispensable for the maintenance of electrochemical gradients across the plasma membrane and herein neuronal excitability. The widely recognized pump function of NKA closely depends on its unique structure features and conformational changes upon binding of specific ions. Various Na+-dependent secondary transport systems are rigorously controlled by the ionic gradients generated by NKA and are essential for multiple physiological processes. In addition, roles of NKA as a signal transducer have also been unveiled nowadays. Plethora of signaling cascades are defined including Src-Ras-MAPK signaling, IP3R-mediated calcium oscillation, inflammation, and autophagy though most underlying mechanisms remain elusive. Ischemic stroke occurs when the blood flow carrying nutrients and oxygen into the brain is disrupted by blood clots, which is manifested by excitotoxicity, oxidative stress, inflammation, etc. The protective effect of NKA against ischemic stress is emerging gradually with the application of specific NKA inhibitor. However, NKA-related research is limited due to the opposite effects caused by NKA inhibitor at lower doses. The present review focuses on the recent progression involving different aspects about NKA in cellular homeostasis to present an in-depth understanding of this unique protein. Moreover, essential roles of NKA in ischemic pathology are discussed to provide a platform and bright future for the improvement in clinical research on ischemic stroke.
Collapse
|
5
|
Abstract
The sodium pump (Na+, K+-ATPase, NKA) is vital for animal cells, as it actively maintains Na+ and K+ electrochemical gradients across the cell membrane. It is a target of cardiotonic steroids (CTSs) such as ouabain and digoxin. As CTSs are almost unique strong inhibitors specific to NKA, a wide range of derivatives has been developed for potential therapeutic use. Several crystal structures have been published for NKA-CTS complexes, but they fail to explain the largely different inhibitory properties of the various CTSs. For instance, although CTSs are thought to inhibit ATPase activity by binding to NKA in the E2P state, we do not know if large conformational changes accompany binding, as no crystal structure is available for the E2P state free of CTS. Here, we describe crystal structures of the BeF3 - complex of NKA representing the E2P ground state and then eight crystal structures of seven CTSs, including rostafuroxin and istaroxime, two new members under clinical trials, in complex with NKA in the E2P state. The conformations of NKA are virtually identical in all complexes with and without CTSs, showing that CTSs bind to a preformed cavity in NKA. By comparing the inhibitory potency of the CTSs measured under four different conditions, we elucidate how different structural features of the CTSs result in different inhibitory properties. The crystal structures also explain K+-antagonism and suggest a route to isoform specific CTSs.
Collapse
|
6
|
Taverner AM, Yang L, Barile ZJ, Lin B, Peng J, Pinharanda AP, Rao AS, Roland BP, Talsma AD, Wei D, Petschenka G, Palladino MJ, Andolfatto P. Adaptive substitutions underlying cardiac glycoside insensitivity in insects exhibit epistasis in vivo. eLife 2019; 8:48224. [PMID: 31453806 PMCID: PMC6733596 DOI: 10.7554/elife.48224] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/24/2019] [Indexed: 01/20/2023] Open
Abstract
Predicting how species will respond to selection pressures requires understanding the factors that constrain their evolution. We use genome engineering of Drosophila to investigate constraints on the repeated evolution of unrelated herbivorous insects to toxic cardiac glycosides, which primarily occurs via a small subset of possible functionally-relevant substitutions to Na+,K+-ATPase. Surprisingly, we find that frequently observed adaptive substitutions at two sites, 111 and 122, are lethal when homozygous and adult heterozygotes exhibit dominant neural dysfunction. We identify a phylogenetically correlated substitution, A119S, that partially ameliorates the deleterious effects of substitutions at 111 and 122. Despite contributing little to cardiac glycoside-insensitivity in vitro, A119S, like substitutions at 111 and 122, substantially increases adult survivorship upon cardiac glycoside exposure. Our results demonstrate the importance of epistasis in constraining adaptive paths. Moreover, by revealing distinct effects of substitutions in vitro and in vivo, our results underscore the importance of evaluating the fitness of adaptive substitutions and their interactions in whole organisms.
Collapse
Affiliation(s)
- Andrew M Taverner
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, United States
| | - Lu Yang
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, United States
| | - Zachary J Barile
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, United States.,Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Becky Lin
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, United States.,Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Julie Peng
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, United States
| | - Ana P Pinharanda
- Department of Biological Sciences, Columbia University, New York, United States
| | - Arya S Rao
- Department of Biological Sciences, Columbia University, New York, United States
| | - Bartholomew P Roland
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, United States.,Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Aaron D Talsma
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, United States.,Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Daniel Wei
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, United States.,Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Georg Petschenka
- Institute for Insect Biotechnology, Justus-Liebig-Universität Gießen, Hesse, Germany
| | - Michael J Palladino
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, United States.,Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Peter Andolfatto
- Department of Biological Sciences, Columbia University, New York, United States
| |
Collapse
|
7
|
Yang L, Ravikanthachari N, Mariño-Pérez R, Deshmukh R, Wu M, Rosenstein A, Kunte K, Song H, Andolfatto P. Predictability in the evolution of Orthopteran cardenolide insensitivity. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180246. [PMID: 31154978 DOI: 10.1098/rstb.2018.0246] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The repeated evolutionary specialization of distantly related insects to cardenolide-containing host plants provides a stunning example of parallel adaptation. Hundreds of herbivorous insect species have independently evolved insensitivity to cardenolides, which are potent inhibitors of the alpha-subunit of Na+,K+-ATPase (ATPα). Previous studies investigating ATPα-mediated cardenolide insensitivity in five insect orders have revealed remarkably high levels of parallelism in the evolution of this trait, including the frequent occurrence of parallel amino acid substitutions at two sites and recurrent episodes of duplication followed by neo-functionalization. Here we add data for a sixth insect order, Orthoptera, which includes an ancient group of highly aposematic cardenolide-sequestering grasshoppers in the family Pyrgomorphidae. We find that Orthopterans exhibit largely predictable patterns of evolution of insensitivity established by sampling other insect orders. Taken together the data lend further support to the proposal that negative pleiotropic constraints are a key determinant in the evolution of cardenolide insensitivity in insects. Furthermore, analysis of our expanded taxonomic survey implicates positive selection acting on site 111 of cardenolide-sequestering species with a single-copy of ATPα, and sites 115, 118 and 122 in lineages with neo-functionalized duplicate copies, all of which are sites of frequent parallel amino acid substitution. This article is part of the theme issue 'Convergent evolution in the genomics era: new insights and directions'.
Collapse
Affiliation(s)
- Lu Yang
- 1 Department of Ecology and Evolutionary Biology, Princeton University , Princeton, NJ 08544 , USA
| | - Nitin Ravikanthachari
- 2 National Centre for Biological Sciences, Tata Institute of Fundamental Research , Bengaluru , India
| | - Ricardo Mariño-Pérez
- 3 Department of Entomology, Texas A&M University , College Station, TX 77843 , USA
| | - Riddhi Deshmukh
- 2 National Centre for Biological Sciences, Tata Institute of Fundamental Research , Bengaluru , India
| | - Mariana Wu
- 1 Department of Ecology and Evolutionary Biology, Princeton University , Princeton, NJ 08544 , USA
| | - Adam Rosenstein
- 1 Department of Ecology and Evolutionary Biology, Princeton University , Princeton, NJ 08544 , USA
| | - Krushnamegh Kunte
- 2 National Centre for Biological Sciences, Tata Institute of Fundamental Research , Bengaluru , India
| | - Hojun Song
- 3 Department of Entomology, Texas A&M University , College Station, TX 77843 , USA
| | - Peter Andolfatto
- 4 Department of Biological Sciences, Columbia University , New York, NY 10027 , USA
| |
Collapse
|
8
|
Coleman DT, Gray AL, Stephens CA, Scott ML, Cardelli JA. Repurposed drug screen identifies cardiac glycosides as inhibitors of TGF-β-induced cancer-associated fibroblast differentiation. Oncotarget 2017; 7:32200-9. [PMID: 27058757 PMCID: PMC5078007 DOI: 10.18632/oncotarget.8609] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/14/2016] [Indexed: 12/22/2022] Open
Abstract
The tumor microenvironment, primarily composed of myofibroblasts, directly influences the progression of solid tumors. Through secretion of growth factors, extracellular matrix deposition, and contractile mechanotransduction, myofibroblasts, or cancer-associated fibroblasts (CAFs), support angiogenesis and cancer cell invasion and metastasis. The differentiation of fibroblasts to CAFs is primarily induced by TGF-β from cancer cells. To discover agents capable of blocking CAF differentiation, we developed a high content immunofluorescence-based assay to screen repurposed chemical libraries utilizing fibronectin expression as an initial CAF marker. Screening of the Prestwick chemical library and NIH Clinical Collection repurposed drug library, totaling over 1700 compounds, identified cardiac glycosides as particularly potent CAF blocking agents. Cardiac glycosides are traditionally used to regulate intracellular calcium by inhibiting the Na+/K+ ATPase to control cardiac contractility. Herein, we report that multiple cardiac glycoside compounds, including digoxin, are able to inhibit TGF-β-induced fibronectin expression at low nanomolar concentrations without undesirable cell toxicity. We found this inhibition to hold true for multiple fibroblast cell lines. Using real-time qPCR, we determined that digoxin prevented induction of multiple CAF markers. Furthermore, we report that digoxin is able to prevent TGF-β-induced fibroblast contraction of extracellular matrix, a major phenotypic consequence of CAF differentiation. Assessing the mechanism of inhibition, we found digoxin reduced SMAD promoter activity downstream of TGF-β, and we provide data that the effect is through inhibition of its known target, the Na+/K+ ATPase. These findings support a critical role for calcium signaling during CAF differentiation and highlight a novel, repurposable modality for cancer therapy.
Collapse
Affiliation(s)
- David T Coleman
- Louisiana State University Health Sciences Center, Feist-Weiller Cancer Center, Shreveport, LA, USA
| | - Alana L Gray
- Louisiana State University Health Sciences Center, Feist-Weiller Cancer Center, Shreveport, LA, USA
| | - Charles A Stephens
- Louisiana State University Health Sciences Center, Feist-Weiller Cancer Center, Shreveport, LA, USA
| | - Matthew L Scott
- Louisiana State University Health Sciences Center, Feist-Weiller Cancer Center, Shreveport, LA, USA
| | - James A Cardelli
- Louisiana State University Health Sciences Center, Feist-Weiller Cancer Center, Shreveport, LA, USA
| |
Collapse
|
9
|
Structures and characterization of digoxin- and bufalin-bound Na+,K+-ATPase compared with the ouabain-bound complex. Proc Natl Acad Sci U S A 2015; 112:1755-60. [PMID: 25624492 DOI: 10.1073/pnas.1422997112] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cardiotonic steroids (CTSs) are specific and potent inhibitors of the Na(+),K(+)-ATPase, with highest affinity to the phosphoenzyme (E2P) forms. CTSs are comprised of a steroid core, which can be glycosylated, and a varying number of substituents, including a five- or six-membered lactone. These functionalities have specific influence on the binding properties. We report crystal structures of the Na(+),K(+)-ATPase in the E2P form in complex with bufalin (a nonglycosylated CTS with a six-membered lactone) and digoxin (a trisaccharide-conjugated CTS with a five-membered lactone) and compare their characteristics and binding kinetics with the previously described E2P-ouabain complex to derive specific details and the general mechanism of CTS binding and inhibition. CTSs block the extracellular cation exchange pathway, and cation-binding sites I and II are differently occupied: A single Mg(2+) is bound in site II of the digoxin and ouabain complexes, whereas both sites are occupied by K(+) in the E2P-bufalin complex. In all complexes, αM4 adopts a wound form, characteristic for the E2P state and favorable for high-affinity CTS binding. We conclude that the occupants of the cation-binding site and the type of the lactone substituent determine the arrangement of αM4 and hypothesize that winding/unwinding of αM4 represents a trigger for high-affinity CTS binding. We find that the level of glycosylation affects the depth of CTS binding and that the steroid core substituents fine tune the configuration of transmembrane helices αM1-2.
Collapse
|
10
|
Zhang LN, Sun YJ, Wang LX, Gao ZB. Glutamate Transporters/Na(+), K(+)-ATPase Involving in the Neuroprotective Effect as a Potential Regulatory Target of Glutamate Uptake. Mol Neurobiol 2015; 53:1124-1131. [PMID: 25586061 DOI: 10.1007/s12035-014-9071-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 12/29/2014] [Indexed: 02/05/2023]
Abstract
The glutamate (Glu) transporters GLAST and GLT-1, as the two most important transporters in brain tissue, transport Glu from the extracellular space into the cell protecting against Glu toxicity. Furthermore, GLAST and GLT-1 are sodium-dependent Glu transporters (GluTs) that rely on sodium and potassium gradients generated principally by Na(+), K(+)-ATPase to generate ion gradients that drive Glu uptake. There is an interaction between Na(+), K(+)-ATPase and GluTs to modulate Glu uptake, and Na(+), K(+)-ATPase α, β or γ subunit can be directly coupled to GluTs, co-localizing with GLAST or GLT-1 in vivo to form a macromolecular complex and operate as a functional unit to regulate glutamatergic neurotransmission. Therefore, GluTs/Na(+), K(+)-ATPase may be involved in the neuroprotective effect as a potential regulatory target of Glu uptake in neurodegenerative diseases induced by Glu-mediated neurotoxicity as the final common pathway.
Collapse
Affiliation(s)
- Li-Nan Zhang
- Department of Pharmacy, Hebei University of Science and Technology, 70 Yuhua East Road, Shijiazhuang, Hebei, 050018, People's Republic of China
| | - Yong-Jun Sun
- Department of Pharmacy, Hebei University of Science and Technology, 70 Yuhua East Road, Shijiazhuang, Hebei, 050018, People's Republic of China
| | - Li-Xue Wang
- Cadre Ward, Capital Medical University Electric Power Teaching Hospital, Compound A1, Taiping Bridge Xili, Beijing, 100073, People's Republic of China
| | - Zi-Bin Gao
- Department of Pharmacy, Hebei University of Science and Technology, 70 Yuhua East Road, Shijiazhuang, Hebei, 050018, People's Republic of China. .,State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, 70 Yuhua East Road, Shijiazhuang, 050018, People's Republic of China.
| |
Collapse
|
11
|
Leone FA, Lucena MN, Rezende LA, Garçon DP, Pinto MR, Mantelatto FL, McNamara JC. A kinetic characterization of (Na+, K+)-ATPase activity in the gills of the pelagic seabob shrimp Xiphopenaeus kroyeri (Decapoda, Penaeidae). J Membr Biol 2014; 248:257-72. [PMID: 25534346 DOI: 10.1007/s00232-014-9765-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 12/04/2014] [Indexed: 10/24/2022]
Abstract
We characterize the kinetic properties of a gill (Na(+), K(+))-ATPase from the pelagic marine seabob Xiphopenaeus kroyeri. Sucrose density gradient centrifugation revealed membrane fractions distributed mainly into a heavy fraction showing considerable (Na(+), K(+))-ATPase activity, but also containing mitochondrial F0F1- and Na(+)- and V-ATPases. Western blot analysis identified a single immunoreactive band against the (Na(+), K(+))-ATPase α-subunit with an Mr of ≈ 110 kDa. The α-subunit was immunolocalized to the intralamellar septum of the gill lamellae. The (Na(+), K(+))-ATPase hydrolyzed ATP obeying Michaelis-Menten kinetics with VM = 109.5 ± 3.2 nmol Pi min(-1) mg(-1) and KM = 0.03 ± 0.003 mmol L(-1). Mg(2+) (VM = 109.8 ± 2.1 nmol Pi min(-1 )mg(-1), K0.5 = 0.60 ± 0.03 mmol L(-1)), Na(+) (VM = 117.6 ± 3.5 nmol Pi min(-1 ) mg(-1), K0.5 = 5.36 ± 0.14 mmol L(-1)), K(+) (VM = 112.9 ± 1.4 nmol Pi min(-1 )mg(-1), K0.5 = 1.32 ± 0.08 mmol L(-1)), and NH4 (+) (VM = 200.8 ± 7.1 nmol Pi min(-1 )mg(-1), K0.5 = 2.70 ± 0.04 mmol L(-1)) stimulated (Na(+), K(+))-ATPase activity following site-site interactions. K(+) plus NH4 (+) does not synergistically stimulate (Na(+), K(+))-ATPase activity, although each ion modulates affinity of the other. The enzyme exhibits a single site for K(+) binding that can be occupied by NH4 (+), stimulating the enzyme. Ouabain (KI = 84.0 ± 2.1 µmol L(-1)) and orthovanadate (KI = 0.157 ± 0.001 µmol L(-1)) inhibited total ATPase activity by ≈ 50 and ≈ 44 %, respectively. Ouabain inhibition increases ≈ 80 % in the presence of NH4 (+) with a threefold lower KI, suggesting that NH4 (+) is likely transported as a K(+) congener.
Collapse
Affiliation(s)
- Francisco Assis Leone
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, SP, 14040-901, Brasil,
| | | | | | | | | | | | | |
Collapse
|
12
|
Rahman MM, Park BY. Na, K-ATPase β2 isoform (atp1b2) expressed in the retina of Xenopus. J Biomed Res 2014. [DOI: 10.12729/jbr.2014.15.4.194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
13
|
Weigand KM, Laursen M, Swarts HGP, Engwerda AHJ, Prüfert C, Sandrock J, Nissen P, Fedosova NU, Russel FGM, Koenderink JB. Na(+),K(+)-ATPase isoform selectivity for digitalis-like compounds is determined by two amino acids in the first extracellular loop. Chem Res Toxicol 2014; 27:2082-92. [PMID: 25361285 DOI: 10.1021/tx500290k] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Digitalis-like compounds (DLCs) comprise a diverse group of molecules characterized by a cis-trans-cis ring-fused steroid core linked to a lactone. They have been used in the treatment of different medical problems including heart failure, where their inotropic effect on heart muscle is attributed to potent Na(+),K(+)-ATPase inhibition. Their application as drugs, however, has declined in recent past years due to their small safety margin. Since human Na(+),K(+)-ATPase is represented by four different isoforms expressed in a tissue-specific manner, one of the possibilities to improve the therapeutic index of DLCs is to exploit and amend their isoform selectivity. Here, we aimed to reveal the determinants of selectivity of the ubiquitously expressed α1 isoform and the more restricted α2 isoform toward several well-known DLCs and their hydrogenated forms. Using baculovirus to express various mutants of the α2 isoform, we were able to link residues Met(119) and Ser(124) to differences in affinity between the α1 and α2 isoforms to ouabain, dihydro-ouabain, digoxin, and dihydro-digoxin. We speculate that the interactions between these amino acids and DLCs affect the initial binding of these DLCs. Also, we observed isoform selectivity for DLCs containing no sugar groups.
Collapse
Affiliation(s)
- Karl M Weigand
- Departments of †Pharmacology and Toxicology and ‡Biochemistry, Radboud University Medical Center , P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Gritz SM, Radcliffe RA. Genetic effects of ATP1A2 in familial hemiplegic migraine type II and animal models. Hum Genomics 2013; 7:8. [PMID: 23561701 PMCID: PMC3639839 DOI: 10.1186/1479-7364-7-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 02/26/2013] [Indexed: 12/19/2022] Open
Abstract
Na+/K+-ATPase alpha 2 (Atp1a2) is an integral plasma membrane protein belonging to the P-type ATPase family that is responsible for maintaining the sodium (Na+) and potassium (K+) gradients across cellular membranes with hydrolysis of ATP. Atp1a2 contains two subunits, alpha and beta, with each having various isoforms and differential tissue distribution. In humans, mutations in ATP1A2 are associated with a rare form of hereditary migraines with aura known as familial hemiplegic migraine type II. Genetic studies in mice have revealed other neurological effects of Atp1a2 in mice including anxiety, fear, and learning and motor function disorders. This paper reviews the recent findings in the literature concerning Atp1a2.
Collapse
Affiliation(s)
- Stephanie M Gritz
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | | |
Collapse
|
15
|
|
16
|
Rossini GP, Bigiani A. Palytoxin action on the Na(+),K(+)-ATPase and the disruption of ion equilibria in biological systems. Toxicon 2010; 57:429-39. [PMID: 20932855 DOI: 10.1016/j.toxicon.2010.09.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 09/03/2010] [Accepted: 09/24/2010] [Indexed: 11/29/2022]
Abstract
Palytoxin-group toxins (PlTX) exert their potent biological activity by altering mechanisms of ion homeostasis in excitable and non-excitable tissues. This review will describe major aspects that led to the relatively early identification of the Na(+),K(+)-ATPase as the molecular target and receptor of the toxin in sensitive systems. The importance of this pump in the normal functioning of animal cells has driven extensive investigative efforts. The recognized molecular mechanism of action of PlTX involves its binding to the extracellular portion of alpha subunit of this plasma membrane protein, which converts an enzyme carrying ions against their concentration gradients at the expense of chemical energy (ATP) into a non-selective cation channel, allowing passive flow of ions following their concentration gradients. More recent findings have indicated that PlTX would interfere with the normal strict coupling between inner and outer gates of the pump controlling the ion access to the Na(+),K(+)-ATPase, allowing the gates to be simultaneously open. The ability of PlTX to make internal portions of the Na(+),K(+)-ATPase accessible to relatively large molecules has been exploited to characterize the structure-function relationship of the pump, leading to a better understanding of its ion translocation pathway. Thus, forty years from the isolation of this potent marine biotoxin, a considerable understanding of its mode of action and of its potential as a research tool have been achieved and are the basis for promising future advancement in the characterization of biological systems and their alteration by PlTX.
Collapse
Affiliation(s)
- Gian Paolo Rossini
- Dipartimento di Scienze Biomediche, Università di Modena e Reggio Emilia, Via G. Campi 287, I-41125 Modena, Italy.
| | | |
Collapse
|
17
|
Abstract
Deactivation of glutamatergic signaling in the brain is mediated by glutamate uptake into glia and neurons by glutamate transporters. Glutamate transporters are sodium-dependent proteins that putatively rely indirectly on Na,K-ATPases to generate ion gradients that drive transmitter uptake. Based on anatomical colocalization, mutual sodium dependency, and the inhibitory effects of the Na,K-ATPase inhibitor ouabain on glutamate transporter activity, we postulated that glutamate transporters are directly coupled to Na,K-ATPase and that Na,K-ATPase is an essential modulator of glutamate uptake. Na,K-ATPase was purified from rat cerebellum by tandem anion exchange and ouabain affinity chromatography, and the cohort of associated proteins was characterized by mass spectrometry. The alpha1-alpha 3 subunits of Na,K-ATPase were detected, as were the glutamate transporters GLAST and GLT-1, demonstrating that glutamate transporters copurify with Na,K-ATPases. The link between glutamate transporters and Na,K-ATPase was further established by coimmunoprecipitation and colocalization. Analysis of the regulation of glutamate transporter and Na,K-ATPase activities was assessed using [(3)H]D-aspartate, [(3)H]L-glutamate, and rubidium-86 uptake into synaptosomes and cultured astrocytes. In synaptosomes, ouabain produced a dose-dependent inhibition of glutamate transporter and Na,K-ATPase activities, whereas in astrocytes, ouabain showed a bimodal effect whereby glutamate transporter activity was stimulated at 1 microm ouabain and inhibited at higher concentrations. The effects of protein kinase inhibitors on [(3)H]D-aspartate uptake indicated the selective involvement of Src kinases, which are probably a component of the Na,K-ATPase/glutamate transporter complex. These findings demonstrate that glutamate transporters and Na,K-ATPases are part of the same macromolecular complexes and operate as a functional unit to regulate glutamatergic neurotransmission.
Collapse
|
18
|
Crystal structure of the sodium-potassium pump (Na+,K+-ATPase) with bound potassium and ouabain. Proc Natl Acad Sci U S A 2009; 106:13742-7. [PMID: 19666591 DOI: 10.1073/pnas.0907054106] [Citation(s) in RCA: 254] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The sodium-potassium pump (Na(+),K(+)-ATPase) is responsible for establishing Na(+) and K(+) concentration gradients across the plasma membrane and therefore plays an essential role in, for instance, generating action potentials. Cardiac glycosides, prescribed for congestive heart failure for more than 2 centuries, are efficient inhibitors of this ATPase. Here we describe a crystal structure of Na(+),K(+)-ATPase with bound ouabain, a representative cardiac glycoside, at 2.8 A resolution in a state analogous to E2.2K(+).Pi. Ouabain is deeply inserted into the transmembrane domain with the lactone ring very close to the bound K(+), in marked contrast to previous models. Due to antagonism between ouabain and K(+), the structure represents a low-affinity ouabain-bound state. Yet, most of the mutagenesis data obtained with the high-affinity state are readily explained by the present crystal structure, indicating that the binding site for ouabain is essentially the same. According to a homology model for the high affinity state, it is a closure of the binding cavity that confers a high affinity.
Collapse
|
19
|
Na⁺,K⁺-ATPase as the Target Enzyme for Organic and Inorganic Compounds. SENSORS 2008; 8:8321-8360. [PMID: 27873990 PMCID: PMC3791021 DOI: 10.3390/s8128321] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2008] [Revised: 11/09/2008] [Accepted: 12/11/2008] [Indexed: 01/16/2023]
Abstract
This paper gives an overview of the literature data concerning specific and non specific inhibitors of Na+,K+-ATPase receptor. The immobilization approaches developed to improve the rather low time and temperature stability of Na+,K+-ATPase, as well to preserve the enzyme properties were overviewed. The functional immobilization of Na+,K+-ATPase receptor as the target, with preservation of the full functional protein activity and access of various substances to an optimum number of binding sites under controlled conditions in the combination with high sensitive technology for the detection of enzyme activity is the basis for application of this enzyme in medical, pharmaceutical and environmental research.
Collapse
|
20
|
Capendeguy O, Iwaszkiewicz J, Michielin O, Horisberger JD. The Fourth Extracellular Loop of the α Subunit of Na,K-ATPase. J Biol Chem 2008; 283:27850-27858. [DOI: 10.1074/jbc.m802194200] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
21
|
Schoner W, Scheiner-Bobis G. Role of endogenous cardiotonic steroids in sodium homeostasis. Nephrol Dial Transplant 2008; 23:2723-9. [PMID: 18556748 DOI: 10.1093/ndt/gfn325] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
22
|
Weiss M. Mechanistic modeling of digoxin distribution kinetics incorporating slow tissue binding. Eur J Pharm Sci 2006; 30:256-63. [PMID: 17194579 DOI: 10.1016/j.ejps.2006.11.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2006] [Accepted: 11/15/2006] [Indexed: 11/17/2022]
Abstract
This study aims to develop a mechanistic pharmacokinetic model that accounts for the kinetics of tissue binding in order to evaluate the effect of slow binding of digoxin to skeletal muscular Na(+)/K(+)-ATPase in humans. The approach is based on a minimal circulatory model with a systemic transit time density function that accounts for vascular mixing, transcapillary permeation and extravascular binding of the drug. The model parameters were estimated using previously published disposition data of digoxin in healthy volunteers and physiological distribution volumes taken from the literature. A time constant of the binding process of 34min was estimated indicating that receptor binding and not permeation clearance is the rate-limiting step of the distribution process. Model simulations suggest that up- or downregulation of sodium pumps, typically observed under physiological or pathophysiological conditions, could be detected with this method. The model allows a quantitative prediction of the effect of changes in skeletal muscular sodium pump activity on plasma levels of digoxin.
Collapse
Affiliation(s)
- Michael Weiss
- Section of Pharmacokinetics, Department of Pharmacology, Martin Luther University Halle-Wittenberg, 06097 Halle, Germany.
| |
Collapse
|
23
|
Artigas P, Gadsby DC. Ouabain affinity determining residues lie close to the Na/K pump ion pathway. Proc Natl Acad Sci U S A 2006; 103:12613-8. [PMID: 16894161 PMCID: PMC1567927 DOI: 10.1073/pnas.0602720103] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Na/K pump establishes essential ion concentration gradients across animal cell membranes. Cardiotonic steroids, such as ouabain, are specific inhibitors of the Na/K pump. We exploited the marine toxin, palytoxin, to probe both the ion translocation pathway through the Na/K pump and the site of its interaction with ouabain. Palytoxin uncouples the pump's gates, which normally open strictly alternately, thus allowing both gates to sometimes be open, so transforming the pump into an ion channel. Palytoxin therefore permits electrophysiological analysis of even a single Na/K pump. We used outside-out patch recording of Xenopus alpha1beta3 Na/K pumps, which were made ouabain-resistant by point mutation, after expressing them in Xenopus oocytes. Endogenous, ouabain-sensitive, Xenopus alpha1beta3 Na/K pumps were silenced by continuous exposure to ouabain. We found that side-chain charge of two residues at either end of the alpha subunit's first extracellular loop, known to make a major contribution to ouabain affinity, strongly influenced conductance of single palytoxin-bound pump-channels by an electrostatic mechanism. The effects were mimicked by modification of cysteines introduced at those two positions with variously charged methanethiosulfonate reagents. The consequences of these modifications demonstrate that both residues lie in a wide vestibule near the mouth of the pump's ion pathway. Bound ouabain protects the site with the strongest influence on conductance from methanethiosulfonate modification, while leaving the site with the weaker influence unprotected. The results suggest a method for mapping the footprint of bound cardiotonic steroid on the extracellular surface of the Na/K pump.
Collapse
Affiliation(s)
- Pablo Artigas
- Laboratory of Cardiac/Membrane Physiology, The Rockefeller University, New York, NY 10021
| | - David C. Gadsby
- Laboratory of Cardiac/Membrane Physiology, The Rockefeller University, New York, NY 10021
- *To whom correspondence should be addressed. E-mail:
| |
Collapse
|
24
|
Kotova O, Al-Khalili L, Talia S, Hooke C, Fedorova OV, Bagrov AY, Chibalin AV. Cardiotonic Steroids Stimulate Glycogen Synthesis in Human Skeletal Muscle Cells via a Src- and ERK1/2-dependent Mechanism. J Biol Chem 2006; 281:20085-94. [PMID: 16714287 DOI: 10.1074/jbc.m601577200] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The cardiotonic steroid, ouabain, a specific inhibitor of Na(+),K(+)-ATPase, initiates protein-protein interactions that lead to an increase in growth and proliferation in different cell types. We explored the effects of ouabain on glucose metabolism in human skeletal muscle cells (HSMC) and clarified the mechanisms of ouabain signal transduction. In HSMC, ouabain increased glycogen synthesis in a concentration-dependent manner reaching the maximum at 100 nM. The effect of ouabain was additive to the effect of insulin and was independent of phosphatidylinositol 3-kinase inhibitor LY294002 but was abolished in the presence of a MEK1/2 inhibitor (PD98059) or a Src inhibitor (PP2). Ouabain increased Src-dependent tyrosine phosphorylation of alpha(1)- and alpha(2)-subunits of Na(+),K(+)-ATPase and promoted interaction of alpha(1)- and alpha(2)-subunits with Src, as assessed by co-immunoprecipitation with Src. Phosphorylation of ERK1/2 and GSK3alpha/beta, as well as p90rsk activity, was increased in response to ouabain in HSMC, and these responses were prevented in the presence of PD98059 and PP2. Incubation of HSMC with 100 nM ouabain increased phosphorylation of the alpha-subunits of the Na-pump at a MAPK-specific Thr-Pro motif. Ouabain treatment decreased the surface abundance of alpha(2)-subunit, whereas abundance of the alpha(1)-subunit was unchanged. Marinobufagenin, an endogenous vertebrate bufadienolide cardiotonic steroid, increased glycogen synthesis in HSMC at 10 nM concentration, similarly to 100 nM ouabain. In conclusion, ouabain and marinobufagenin stimulate glycogen synthesis in skeletal muscle. This effect is mediated by activation of a Src-, ERK1/2-, p90rsk-, and GSK3-dependent signaling pathway.
Collapse
Affiliation(s)
- Olga Kotova
- Section of Integrative Physiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
25
|
Suwalsky M, Mennickent S, Norris B, Cárdenas H. The antiepileptic drug phenytoin affects sodium transport in toad epithelium. Comp Biochem Physiol C Toxicol Pharmacol 2006; 142:253-261. [PMID: 16314149 DOI: 10.1016/j.cbpc.2005.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2005] [Revised: 10/09/2005] [Accepted: 10/11/2005] [Indexed: 11/26/2022]
Abstract
The effects of phenytoin on isolated Pleurodema thaul toad skin were investigated. Low (micromolar) concentrations of the antiepileptic agent applied to the outside surface of the toad epithelium increased the electrical parameters (short-circuit current and potential difference) by over 40%, reflecting stimulation of Na(+) transport, whereas higher (millimolar concentrations, outside and inside surface) decreased both electric parameters, the effect being greater at the inside surface (40% and 80% decrease, respectively). The amiloride test showed that the stimulatory effect was accompanied by an increase and the inhibitory effect by a decrease in the sodium electromotive force (ENa). It is concluded that the drug interaction with membrane lipid bilayers might result in a distortion of the lipid-protein interface contributing to disturbance of Na(+) epithelial channel activity. After applying the Na(+)-K(+)-ATPase blocker ouabain and replacing the Na(+) ions in the outer Ringer's solution by choline, it was concluded that both active and passive transport are involved in sodium absorption, although active transport predominates.
Collapse
Affiliation(s)
- Mario Suwalsky
- Faculty of Chemical Sciences, University of Concepción, Casilla 160-C, Concepcion, Chile.
| | - Sigrid Mennickent
- Faculty of Pharmacy, University of Concepción, Casilla 160-C, Concepcion, Chile
| | - Beryl Norris
- Faculty of Biological Sciences, University of Concepción, Casilla 237, Concepcion, Chile
| | - Hernan Cárdenas
- Faculty of Biological Sciences, University of Concepción, Casilla 237, Concepcion, Chile
| |
Collapse
|
26
|
Qiu LY, Krieger E, Schaftenaar G, Swarts HGP, Willems PHGM, De Pont JJHHM, Koenderink JB. Reconstruction of the Complete Ouabain-binding Pocket of Na,K-ATPase in Gastric H,K-ATPase by Substitution of Only Seven Amino Acids. J Biol Chem 2005; 280:32349-55. [PMID: 16051601 DOI: 10.1074/jbc.m505168200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although cardiac glycosides have been used as drugs for more than 2 centuries and their primary target, the sodium pump (Na,K-ATPase), has already been known for 4 decades, their exact binding site is still elusive. In our efforts to define the molecular basis of digitalis glycosides binding we started from the fact that a closely related enzyme, the gastric H,K-ATPase, does not bind glycosides like ouabain. Previously, we showed that a chimera of these two enzymes, in which only the M3-M4 and M5-M6 hairpins were of Na,K-ATPase, bound ouabain with high affinity (Koenderink, J. B., Hermsen, H. P. H., Swarts, H. G. P., Willems, P. H. G. M., and De Pont, J. J. H. H. M. (2000) Proc. Natl. Acad. Sci. U. S. A. 97, 11209-11214). We also demonstrated that only three amino acids (Phe(783), Thr(797), and Asp(804)) present in the M5-M6 hairpin of Na,K-ATPase were sufficient to confer high affinity ouabain binding to a chimera which contained in addition the M3-M4 hairpin of Na,K-ATPase (Qiu, L. Y., Koenderink, J. B., Swarts, H. G., Willems, P. H., and De Pont, J. J. H. H. M. (2003) J. Biol. Chem. 278, 47240-47244). To further pinpoint the ouabain-binding site here we used a chimera-based loss-of-function strategy and identified four amino acids (Glu(312), Val(314), Ile(315), Gly(319)), all present in M4, as being important for ouabain binding. In a final gain-of-function study we showed that a gastric H,K-ATPase that contained Glu(312), Val(314), Ile(315), Gly(319), Phe(783), Thr(797), and Asp(804) of Na,K-ATPase bound ouabain with the same affinity as the native enzyme. Based on the E(2)P crystal structure of Ca(2+)-ATPase we constructed a homology model for the ouabain-binding site of Na,K-ATPase involving all seven amino acids as well as several earlier postulated amino acids.
Collapse
Affiliation(s)
- Li Yan Qiu
- Department of Biochemistry (160), Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
The sodium pump (or Na-K-ATPase) is essential to the function of animal cells. Publication of the related calcium pump (SERCA) structure together with several recent results from a variety of approaches allow us to propose a mechanistic model to answer the question: “How does the sodium pump pump?”
Collapse
Affiliation(s)
- J-D Horisberger
- Department of Pharmacology and Toxicology, University of Lausanne, CH-1005 Lausanne, Switzerland.
| |
Collapse
|
28
|
Abstract
Because nearly all structure/function studies on Na(+)/K(+)-ATPase have been done on enzymes prepared in the presence of SDS, we have studied previously unrecognized consequences of SDS interaction with the enzyme. When the purified membrane-bound kidney enzyme was solubilized with SDS or TDS concentrations just sufficient to cause complete solubilization, but not at concentrations severalfold higher, the enzyme retained quaternary structure, exhibiting alpha,alpha-, alpha,beta-, beta,beta-, and alpha,gamma-associations as detected by chemical cross-linking. The presence of solubilized oligomers was confirmed by sucrose density gradient centrifugation. This solubilized enzyme had no ATPase activity and was not phosphorylated by ATP, but it retained the ability to occlude Rb(+) and Na(+). This, and comparison of cross-linking patterns obtained with different reagents, suggested that the transmembrane domains of the enzyme are more resistant to SDS-induced unfolding than its other domains. These findings (a). indicate that the partially unfolded oligomer(s) retaining partial function is the intermediate in the SDS-induced denaturation of the native membrane enzyme having the minimum oligomeric structure of (alpha,beta,gamma)(2) and (b). suggest potential functions for Na(+)/K(+)-ATPase with intrinsically unfolded domains. Mixtures of solubilized/partially unfolded enzyme and membrane-bound enzyme exhibited cross-linking patterns and Na(+) occlusion capacities different from those of either enzyme species, suggesting that the two interact. Formation of the partially unfolded enzyme during standard purification procedure for the preparation of the membrane-bound enzyme was shown, indicating that it is necessary to ensure the separation of the partially unfolded enzyme from the membrane-bound enzyme to avoid the distortion of the properties of the latter.
Collapse
Affiliation(s)
- Alexander V Ivanov
- Department of Pharmacology, Medical College of Ohio, Toledo, Ohio 43614-5804, USA
| | | | | |
Collapse
|