Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.332
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 89: 583-603, 2020 06 20.
Article in English | MEDLINE | ID: mdl-31874046

ABSTRACT

P-type ATPases are found in all kingdoms of life and constitute a wide range of cation transporters, primarily for H+, Na+, K+, Ca2+, and transition metal ions such as Cu(I), Zn(II), and Cd(II). They have been studied through a wide range of techniques, and research has gained very significant insight on their transport mechanism and regulation. Here, we review the structure, function, and dynamics of P2-ATPases including Ca2+-ATPases and Na,K-ATPase. We highlight mechanisms of functional transitions that are associated with ion exchange on either side of the membrane and how the functional cycle is regulated by interaction partners, autoregulatory domains, and off-cycle states. Finally, we discuss future perspectives based on emerging techniques and insights.


Subject(s)
Adenosine Triphosphate/chemistry , Copper-Transporting ATPases/chemistry , H(+)-K(+)-Exchanging ATPase/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Sodium-Potassium-Exchanging ATPase/chemistry , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Cations, Divalent , Cations, Monovalent , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , H(+)-K(+)-Exchanging ATPase/genetics , H(+)-K(+)-Exchanging ATPase/metabolism , Humans , Ion Transport , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Protons , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Single Molecule Imaging , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Substrate Specificity
2.
Nature ; 583(7815): 314-318, 2020 07.
Article in English | MEDLINE | ID: mdl-32499654

ABSTRACT

Light-driven sodium pumps actively transport small cations across cellular membranes1. These pumps are used by microorganisms to convert light into membrane potential and have become useful optogenetic tools with applications in neuroscience. Although the resting state structures of the prototypical sodium pump Krokinobacter eikastus rhodopsin 2 (KR2) have been solved2,3, it is unclear how structural alterations over time allow sodium to be translocated against a concentration gradient. Here, using the Swiss X-ray Free Electron Laser4, we have collected serial crystallographic data at ten pump-probe delays from femtoseconds to milliseconds. High-resolution structural snapshots throughout the KR2 photocycle show how retinal isomerization is completed on the femtosecond timescale and changes the local structure of the binding pocket in the early nanoseconds. Subsequent rearrangements and deprotonation of the retinal Schiff base open an electrostatic gate in microseconds. Structural and spectroscopic data, in combination with quantum chemical calculations, indicate that a sodium ion binds transiently close to the retinal within one millisecond. In the last structural intermediate, at 20 milliseconds after activation, we identified a potential second sodium-binding site close to the extracellular exit. These results provide direct molecular insight into the dynamics of active cation transport across biological membranes.


Subject(s)
Flavobacteriaceae/chemistry , Rhodopsins, Microbial/chemistry , Rhodopsins, Microbial/radiation effects , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/radiation effects , Binding Sites , Crystallography , Electrons , Ion Transport , Isomerism , Lasers , Protons , Quantum Theory , Retinaldehyde/chemistry , Retinaldehyde/metabolism , Schiff Bases/chemistry , Sodium/metabolism , Spectrum Analysis , Static Electricity , Time Factors
3.
Circ Res ; 132(9): 1127-1140, 2023 04 28.
Article in English | MEDLINE | ID: mdl-36919600

ABSTRACT

BACKGROUND: Extracellular renal interstitial guanosine cyclic 3',5'-monophosphate (cGMP) inhibits renal proximal tubule (RPT) sodium (Na+) reabsorption via Src (Src family kinase) activation. Through which target extracellular cGMP acts to induce natriuresis is unknown. We hypothesized that cGMP binds to the extracellular α1-subunit of NKA (sodium-potassium ATPase) on RPT basolateral membranes to inhibit Na+ transport similar to ouabain-a cardiotonic steroid. METHODS: Urine Na+ excretion was measured in uninephrectomized 12-week-old female Sprague-Dawley rats that received renal interstitial infusions of vehicle (5% dextrose in water), cGMP (18, 36, and 72 µg/kg per minute; 30 minutes each), or cGMP+rostafuroxin (12 ng/kg per minute) or were subjected to pressure-natriuresis±rostafuroxin infusion. Rostafuroxin is a digitoxigenin derivative that displaces ouabain from NKA. RESULTS: Renal interstitial cGMP and raised renal perfusion pressure induced natriuresis and increased phosphorylated SrcTyr416 and Erk 1/2 (extracellular signal-regulated protein kinase 1/2)Thr202/Tyr204; these responses were abolished with rostafuroxin coinfusion. To assess cGMP binding to NKA, we performed competitive binding studies with isolated rat RPTs using bodipy-ouabain (2 µM)+cGMP (10 µM) or rostafuroxin (10 µM) and 8-biotin-11-cGMP (2 µM)+ouabain (10 µM) or rostafuroxin (10 µM). cGMP or rostafuroxin reduced bodipy-ouabain fluorescence intensity, and ouabain or rostafuroxin reduced 8-biotin-11-cGMP staining. We cross-linked isolated rat RPTs with 4-N3-PET-8-biotin-11-cGMP (2 µM); 8-N3-6-biotin-10-cAMP served as negative control. Precipitation with streptavidin beads followed by immunoblot analysis showed that RPTs after cross-linking with 4-N3-PET-8-biotin-11-cGMP exhibited a significantly stronger signal for NKA than non-cross-linked samples and cross-linked or non-cross-linked 8-N3-6-biotin-10-cAMP RPTs. Ouabain (10 µM) reduced NKA in cross-linked 4-N3-PET-8-biotin-11-cGMP RPTs confirming fluorescence staining. 4-N3-PET-8-biotin-11-cGMP cross-linked samples were separated by SDS gel electrophoresis and slices corresponding to NKA molecular weight excised and processed for mass spectrometry. NKA was the second most abundant protein with 50 unique NKA peptides covering 47% of amino acids in NKA. Molecular modeling demonstrated a potential cGMP docking site in the ouabain-binding pocket of NKA. CONCLUSIONS: cGMP can bind to NKA and thereby mediate natriuresis.


Subject(s)
Cyclic GMP , Natriuresis , Sodium-Potassium-Exchanging ATPase , Animals , Female , Rats , Adenosine Triphosphatases/metabolism , Biotin/metabolism , Cyclic GMP/chemistry , Cyclic GMP/metabolism , Natriuresis/physiology , Ouabain/pharmacology , Potassium/metabolism , Rats, Sprague-Dawley , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/metabolism
4.
Proc Natl Acad Sci U S A ; 119(15): e2123226119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35380894

ABSTRACT

Cryoelectron microscopy (cryo-EM) was applied to Na+,K+-ATPase (NKA) to determine the structures of two E2P states, one (E2PATP) formed by ATP and Mg2+ in the forward reaction, and the other (E2PPi) formed by inorganic phosphate (Pi) and Mg2+ in the backward reaction, with and without ouabain or istaroxime, representatives of classical and new-generation cardiotonic steroids (CTSs). These two E2P states exhibit different biochemical properties. In particular, K+-sensitive acceleration of the dephosphorylation reaction is not observed with E2PPi, attributed to the presence of a Mg2+ ion in the transmembrane cation binding sites. The cryo-EM structures of NKA demonstrate that the two E2P structures are nearly identical but Mg2+ in the transmembrane binding cavity is identified only in E2PPi, corroborating the idea that it should be denoted as E2PPi·Mg2+. We can now explain why the absence of transmembrane Mg2+ in E2PATP confers the K+ sensitivity in dephosphorylation. In addition, we show that ATP bridges the actuator (A) and nucleotide binding (N) domains, stabilizing the E2PATP state; CTS binding causes hardly any changes in the structure of NKA, both in E2PATP and E2PPi·Mg2+, indicating that the binding mechanism is conformational selection; and istaroxime binds to NKA, extending its aminoalkyloxime group deep into the cation binding site. This orientation is upside down compared to that of classical CTSs with respect to the steroid ring. Notably, mobile parts of NKA are resolved substantially better in the electron microscopy (EM) maps than in previous X-ray structures, including sugars sticking out from the ß-subunit and many phospholipid molecules.


Subject(s)
Cardiac Glycosides , Cardiotonic Agents , Etiocholanolone/analogs & derivatives , Ouabain , Sodium-Potassium-Exchanging ATPase , Adenosine Triphosphate/metabolism , Cardiac Glycosides/pharmacology , Cardiotonic Agents/pharmacology , Cations/chemistry , Cations/metabolism , Cryoelectron Microscopy , Etiocholanolone/pharmacology , Magnesium/chemistry , Magnesium/metabolism , Ouabain/pharmacology , Protein Domains , Sodium/chemistry , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/metabolism
5.
J Biol Chem ; 299(2): 102811, 2023 02.
Article in English | MEDLINE | ID: mdl-36539036

ABSTRACT

The Na+/K+-ATPase is an integral plasma membrane glycoprotein of all animal cells that couples the exchange of intracellular Na+ for extracellular K+ to the hydrolysis of ATP. The asymmetric distribution of Na+ and K+ is essential for cellular life and constitutes the physical basis of a series of fundamental biological phenomena. The pumping mechanism is explained by the Albers-Post model. It involves the presence of gates alternatively exposing Na+/K+-ATPase transport sites to the intracellular and extracellular sides and includes occluded states in which both gates are simultaneously closed. Unlike for K+, information is lacking about Na+-occluded intermediates, as occluded Na+ was only detected in states incapable of performing a catalytic cycle, including two Na+-containing crystallographic structures. The current knowledge is that intracellular Na+ must bind to the transport sites and become occluded upon phosphorylation by ATP to be transported to the extracellular medium. Here, taking advantage of epigallocatechin-3-gallate to instantaneously stabilize native Na+-occluded intermediates, we isolated species with tightly bound Na+ in an enzyme able to perform a catalytic cycle, consistent with a genuine occluded state. We found that Na+ becomes spontaneously occluded in the E1 dephosphorylated form of the Na+/K+-ATPase, exhibiting positive interactions between binding sites. In fact, the addition of ATP does not produce an increase in Na+ occlusion as it would have been expected; on the contrary, occluded Na+ transiently decreases, whereas ATP lasts. These results reveal new properties of E1 intermediates of the Albers-Post model for explaining the Na+ transport pathway.


Subject(s)
Biocatalysis , Sodium-Potassium-Exchanging ATPase , Sodium , Animals , Adenosine Triphosphate/metabolism , Cell Membrane/metabolism , Kinetics , Potassium/metabolism , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/metabolism , Ion Transport , Phosphorylation , Cations, Monovalent/metabolism
6.
J Chem Inf Model ; 64(3): 974-982, 2024 02 12.
Article in English | MEDLINE | ID: mdl-38237560

ABSTRACT

Krokinobacter eikastus rhodopsin 2 (KR2) is a typical light-driven sodium pump. Although wild-type KR2 exhibits high Na+ selectivity, mutagenesis performed on the residues constituting the entrance enables permeation of K+ and Cs+, while the underlying mechanism remains elusive. This study presents a comprehensive molecular dynamics investigation, including force field optimization, metadynamics, and alchemical free energy methods, to explore the N61L/G263F mutant of KR2, which exhibits transportability for K+ and Cs+. The introduced Phe263 residue can directly promote ion binding at the entrance through cation-π interactions, while the N61L mutation can enhance ion binding at Phe46 by relieving steric hindrance. These results suggest that cation-π interactions may significantly influence the ion transportability and selectivity of KR2, which can provide important insights for protein engineering and the design of artificial ion transporters.


Subject(s)
Flavobacteriaceae , Molecular Dynamics Simulation , Sodium-Potassium-Exchanging ATPase , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Rhodopsin/chemistry , Rhodopsin/genetics , Rhodopsin/metabolism , Cations/metabolism
7.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Article in English | MEDLINE | ID: mdl-33597302

ABSTRACT

Cellular survival requires the ion gradients built by the Na+/K+ pump, an ATPase that alternates between two major conformations (E1 and E2). Here we use state-specific engineered-disulfide cross-linking to demonstrate that transmembrane segment 2 (M2) of the pump's α-subunit moves in directions that are inconsistent with distances observed in existing crystal structures of the Na+/K+ pump in E1 and E2. We characterize this movement with voltage-clamp fluorometry in single-cysteine mutants. Most mutants in the M1-M2 loop produced state-dependent fluorescence changes upon labeling with tetramethylrhodamine-6-maleimide (TMRM), which were due to quenching by multiple endogenous tryptophans. To avoid complications arising from multiple potential quenchers, we analyzed quenching of TMRM conjugated to R977C (in the static M9-M10 loop) by tryptophans introduced, one at a time, in M1-M2. This approach showed that tryptophans introduced in M2 quench TMRM only in E2, with D126W and L130W on the same helix producing the largest fluorescence changes. These observations indicate that M2 moves outward as Na+ is deoccluded from the E1 conformation, a mechanism consistent with cross-linking results and with proposals for other P-type 2 ATPases.


Subject(s)
Cysteine/chemistry , Oocytes/physiology , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium/metabolism , Animals , Cysteine/genetics , Cysteine/metabolism , Fluorometry , Oocytes/cytology , Protein Conformation , Protein Domains , Sodium-Potassium-Exchanging ATPase/genetics , Xenopus laevis
8.
Proc Natl Acad Sci U S A ; 118(1)2021 01 07.
Article in English | MEDLINE | ID: mdl-33318128

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.


Subject(s)
Cardiac Glycosides/chemistry , Cardiac Glycosides/pharmacology , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium/chemistry , Animals , Biophysical Phenomena , Digoxin/pharmacology , Models, Molecular , Molecular Conformation , Ouabain/pharmacology , Protein Isoforms , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism
9.
Article in English | MEDLINE | ID: mdl-39004301

ABSTRACT

Decapod Crustacea exhibit a marine origin, but many taxa have occupied environments ranging from brackish to fresh water and terrestrial habitats, overcoming their inherent osmotic challenges. Osmotic and ionic regulation is achieved by the gill epithelia, driven by two active ATP-hydrolyzing ion transporters, the basal (Na+, K+)-ATPase and the apical V(H+)-ATPase. The kinetic characteristic of gill (Na+, K+)-ATPase and the mRNA expression of its α subunit have been widely studied in various decapod species under different salinity challenges. However, the evolution of the primary structure has not been explored, especially considering the functional modifications associated with decapod phylogeny. Here, we proposed a model for the topology of the decapod α subunit, identifying the sites and motifs involved in its function and regulation, as well as the patterns of its evolution assuming a decapod phylogeny. We also examined both the amino acid substitutions and their functional implications within the context of biochemical and physiological adaptation. The α-subunit of decapod crustaceans shows greater conservation (∼94% identity) compared to the ß-subunit (∼40%). While the binding sites for ATP and modulators are conserved in the decapod enzyme, the residues involved in the α-ß interaction are only partially conserved. In the phylogenetic context of the complete sequence of (Na+, K+)-ATPase α-subunit, most substitutions appear to be characteristic of the entire group, with specific changes for different subgroups, especially among brachyuran crabs. Interestingly, there was no consistent separation of α-subunit partial sequences related to habitat, suggesting that the convergent evolution for freshwater or terrestrial modes of life is not correlated with similar changes in the enzyme's primary amino acid sequence.


Subject(s)
Amino Acid Sequence , Decapoda , Osmoregulation , Phylogeny , Sodium-Potassium-Exchanging ATPase , Animals , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium-Potassium-Exchanging ATPase/chemistry , Osmoregulation/genetics , Decapoda/genetics , Decapoda/enzymology , Decapoda/physiology , Evolution, Molecular , Gills/metabolism , Gills/enzymology
10.
Int J Mol Sci ; 25(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38892309

ABSTRACT

The sodium pump, or Na+/K+-ATPase (NKA), is an essential enzyme found in the plasma membrane of all animal cells. Its primary role is to transport sodium (Na+) and potassium (K+) ions across the cell membrane, using energy from ATP hydrolysis. This transport creates and maintains an electrochemical gradient, which is crucial for various cellular processes, including cell volume regulation, electrical excitability, and secondary active transport. Although the role of NKA as a pump was discovered and demonstrated several decades ago, it remains the subject of intense research. Current studies aim to delve deeper into several aspects of this molecular entity, such as describing its structure and mode of operation in atomic detail, understanding its molecular and functional diversity, and examining the consequences of its malfunction due to structural alterations. Additionally, researchers are investigating the effects of various substances that amplify or decrease its pumping activity. Beyond its role as a pump, growing evidence indicates that in various cell types, NKA also functions as a receptor for cardiac glycosides like ouabain. This receptor activity triggers the activation of various signaling pathways, producing significant morphological and physiological effects. In this report, we present the results of a comprehensive review of the most outstanding studies of the past five years. We highlight the progress made regarding this new concept of NKA and the various cardiac glycosides that influence it. Furthermore, we emphasize NKA's role in epithelial physiology, particularly its function as a receptor for cardiac glycosides that trigger intracellular signals regulating cell-cell contacts, proliferation, differentiation, and adhesion. We also analyze the role of NKA ß-subunits as cell adhesion molecules in glia and epithelial cells.


Subject(s)
Sodium-Potassium-Exchanging ATPase , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium-Potassium-Exchanging ATPase/chemistry , Animals , Humans , Cell Membrane/metabolism , Signal Transduction , Ouabain/pharmacology , Ouabain/metabolism , Cardiac Glycosides/metabolism , Cardiac Glycosides/pharmacology , Sodium/metabolism
11.
J Biol Chem ; 298(9): 102317, 2022 09.
Article in English | MEDLINE | ID: mdl-35926706

ABSTRACT

The Na+,K+-ATPase generates electrochemical gradients of Na+ and K+ across the plasma membrane via a functional cycle that includes various phosphoenzyme intermediates. However, the structure and function of these intermediates and how metal fluorides mimick them require further investigation. Here, we describe a 4.0 Å resolution crystal structure and functional properties of the pig kidney Na+,K+-ATPase stabilized by the inhibitor beryllium fluoride (denoted E2-BeFx). E2-BeFx is expected to mimic properties of the E2P phosphoenzyme, yet with unknown characteristics of ion and ligand binding. The structure resembles the E2P form obtained by phosphorylation from inorganic phosphate (Pi) and stabilized by cardiotonic steroids, including a low-affinity Mg2+ site near ion binding site II. Our anomalous Fourier analysis of the crystals soaked in Rb+ (a K+ congener) followed by a low-resolution rigid-body refinement (6.9-7.5 Å) revealed preocclusion transitions leading to activation of the dephosphorylation reaction. We show that the Mg2+ location indicates a site of initial K+ recognition and acceptance upon binding to the outward-open E2P state after Na+ release. Furthermore, using binding and activity studies, we find that the BeFx-inhibited enzyme is also able to bind ADP/ATP and Na+. These results relate the E2-BeFx complex to a transient K+- and ADP-sensitive E∗P intermediate of the functional cycle of the Na+,K+-ATPase, prior to E2P.


Subject(s)
Beryllium , Cardiac Glycosides , Fluorides , Kidney , Sodium-Potassium-Exchanging ATPase , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Beryllium/chemistry , Cardiac Glycosides/chemistry , Fluorides/chemistry , Kidney/enzymology , Kinetics , Phosphates/metabolism , Phosphorylation , Protein Domains , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/chemistry , Swine
12.
Nat Rev Mol Cell Biol ; 12(1): 60-70, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21179061

ABSTRACT

Plasma membrane ATPases are primary active transporters of cations that maintain steep concentration gradients. The ion gradients and membrane potentials derived from them form the basis for a range of essential cellular processes, in particular Na(+)-dependent and proton-dependent secondary transport systems that are responsible for uptake and extrusion of metabolites and other ions. The ion gradients are also both directly and indirectly used to control pH homeostasis and to regulate cell volume. The plasma membrane H(+)-ATPase maintains a proton gradient in plants and fungi and the Na(+),K(+)-ATPase maintains a Na(+) and K(+) gradient in animal cells. Structural information provides insight into the function of these two distinct but related P-type pumps.


Subject(s)
Cell Membrane/metabolism , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/metabolism , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Cell Membrane/chemistry , Humans
13.
Chem Pharm Bull (Tokyo) ; 71(2): 154-164, 2023.
Article in English | MEDLINE | ID: mdl-36724978

ABSTRACT

Rhodopsins are transmembrane proteins with retinal chromophores that are involved in photo-energy conversion and photo-signal transduction in diverse organisms. In this study, we newly identified and characterized a rhodopsin from a thermophilic bacterium, Bellilinea sp. Recombinant Escherichia coli cells expressing the rhodopsin showed light-induced alkalization of the medium only in the presence of sodium ions (Na+), and the alkalization signal was enhanced by addition of a protonophore, indicating an outward Na+ pump function across the cellular membrane. Thus, we named the protein Bellilinea Na+-pumping rhodopsin, BeNaR. Of note, its Na+-pumping activity is significantly greater than that of the known Na+-pumping rhodopsin, KR2. We further characterized its photochemical properties as follows: (i) Visible spectroscopy and HPLC revealed that BeNaR has an absorption maximum at 524 nm with predominantly (>96%) the all-trans retinal conformer. (ii) Time-dependent thermal denaturation experiments revealed that BeNaR showed high thermal stability. (iii) The time-resolved flash-photolysis in the nanosecond to millisecond time domains revealed the presence of four kinetically distinctive photointermediates, K, L, M and O. (iv) Mutational analysis revealed that Asp101, which acts as a counterion, and Asp230 around the retinal were essential for the Na+-pumping activity. From the results, we propose a model for the outward Na+-pumping mechanism of BeNaR. The efficient Na+-pumping activity of BeNaR and its high stability make it a useful model both for ion transporters and optogenetics tools.


Subject(s)
Rhodopsin , Sodium-Potassium-Exchanging ATPase , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/metabolism , Rhodopsin/chemistry , Rhodopsin/metabolism , Ion Transport , Bacteria/metabolism , Ions , Sodium/chemistry , Sodium/metabolism , Light
14.
Mol Biol (Mosk) ; 57(6): 1077-1083, 2023.
Article in Russian | MEDLINE | ID: mdl-38062961

ABSTRACT

Melittin, a peptide from bee venom, was found to be able to interact with many proteins, including calmodulin target proteins and ion-transporting P-type ATPases. It is assumed that melittin mimics a protein module involved in protein-protein interactions within cells. Previously, a Na^(+)/K^(+)-ATPase containing the α1 isoform of the catalytic subunit was found to co-precipitate with a protein with a molecular weight of about 70 κDa that interacts with antibodies against melittin by cross immunoprecipitation. In the presence of a specific Na^(+)/K^(+)-ATPase inhibitor (ouabain), the amount of protein with a molecular weight of 70 κDa interacting with Na^(+)/K^(+)-ATPase increases. In order to identify melittin-like protein from murine kidney homogenate, a fraction of melittin-like proteins with a molecular weight of approximately 70 κDa was obtained using affinity chromatography with immobilized antibodies specific to melittin. By mass spectrometry analysis, the obtained protein fraction was found to contain three molecular chaperones of Hsp70 superfamily: mitochondrial mtHsp70 (mortalin), Hsp73, Grp78 (BiP) of endoplasmic reticulum. These data suggest that chaperones from the HSP-70 superfamily contain a melittin-like module.


Subject(s)
Melitten , Sodium-Potassium-Exchanging ATPase , Mice , Animals , Melitten/chemistry , Melitten/metabolism , Melitten/pharmacology , Sodium-Potassium-Exchanging ATPase/chemistry , Molecular Weight , Ouabain/pharmacology , Peptides/metabolism , Molecular Chaperones/metabolism
15.
J Biol Chem ; 296: 100459, 2021.
Article in English | MEDLINE | ID: mdl-33639164

ABSTRACT

The light-driven rhodopsin KR2 transports Na+via the M- and O-states. However, the mechanisms by which the retinal regulates Na+ pumping is unknown, in part because KR2 adopts both pentamer and monomer forms in crystal structures and in part because these structures show differences in the protein conformation near the Schiff base, even when they are of the same intermediate state within the photocycle. A particular open question is the nature of the H-bond networks and protonation state in the active site, including Asp116. Here, we analyze the protonation state and the absorption wavelength for each crystal structure, using a quantum mechanical/molecular mechanical approach. In the pentamer ground state, the calculated absorption wavelength reproduces the experimentally measured absorption wavelength (530 nm). The analysis also shows that ionized Asp116 is stabilized by the H-bond donations of both Ser70 and a cluster of water molecules. The absorption wavelength of 400 nm in the M-state can be best reproduced when the two O atoms of Asp116 interact strongly with the Schiff base, as reported in one of the previous monomer ground state structures. The absorption wavelengths calculated for the two Na+-incorporated O-state structures are consistent with the measured absorption wavelength (∼600 nm), which suggests that two conformations represent the O-state. These results may provide a key to designing enhanced tools in optogenetics.


Subject(s)
Bacterial Proteins/chemistry , Flavobacteriaceae/chemistry , Light , Rhodopsin/chemistry , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium/chemistry , Bacterial Proteins/metabolism , Flavobacteriaceae/metabolism , Protein Domains , Rhodopsin/metabolism , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism
16.
Physiol Rev ; 95(1): 297-340, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25540145

ABSTRACT

Transcription and translation require a high concentration of potassium across the entire tree of life. The conservation of a high intracellular potassium was an absolute requirement for the evolution of life on Earth. This was achieved by the interplay of P- and V-ATPases that can set up electrochemical gradients across the cell membrane, an energetically costly process requiring the synthesis of ATP by F-ATPases. In animals, the control of an extracellular compartment was achieved by the emergence of multicellular organisms able to produce tight epithelial barriers creating a stable extracellular milieu. Finally, the adaptation to a terrestrian environment was achieved by the evolution of distinct regulatory pathways allowing salt and water conservation. In this review we emphasize the critical and dual role of Na(+)-K(+)-ATPase in the control of the ionic composition of the extracellular fluid and the renin-angiotensin-aldosterone system (RAAS) in salt and water conservation in vertebrates. The action of aldosterone on transepithelial sodium transport by activation of the epithelial sodium channel (ENaC) at the apical membrane and that of Na(+)-K(+)-ATPase at the basolateral membrane may have evolved in lungfish before the emergence of tetrapods. Finally, we discuss the implication of RAAS in the origin of the present pandemia of hypertension and its associated cardiovascular diseases.


Subject(s)
Aldosterone/metabolism , Biological Evolution , Epithelial Sodium Channels/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium/metabolism , Animals , Epithelial Sodium Channels/chemistry , Epithelial Sodium Channels/genetics , Genome, Human , Humans , Nephrons/physiology , Signal Transduction/physiology , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/genetics
17.
J Bioenerg Biomembr ; 54(5-6): 241-249, 2022 12.
Article in English | MEDLINE | ID: mdl-36331672

ABSTRACT

Na,K-ATPase is a member of the P-type ATPase family, which transforms the energy of ATP to the transmembrane Na/K gradient that is used to create membrane potential, support the excitability of neurons and myocytes, control pH, and transport substances. The regulation of the Na,K-ATPase function by physiological regulators also comprises a central role in the adaptation of organisms to different conditions. H2O2 is one of the main signaling molecules in redox metabolism and plays important function in cellular physiology. H2O2 also regulates signaling pathways via the specific oxidation of proteins harboring redox-sensitive moieties, like metal centers or cysteine residues, which control their activity. The Na,K-ATPase is redox-sensitive with an "optimal redox potential range," where the reactive oxygen species (ROS), levels beyond this "optimal range" are responsible for enzyme inhibition. Thus reactive oxygen species manifest a hermetic effect, which is expressed by biphasic action; stimulation by low doses and inhibition by high doses. This study was aimed to reveal redox-sensitivity of brain synaptic membrane fractions Na,K-ATPase via H2O2 effects. Different concentrations of H2O2 change the kinetic parameters of the enzyme system for MgATP complex, Na+, and K+ differently. Moreover, H2O2 changes p-chloromercuribenzoic acids (PCMB) affinity. H2O2 targets thiols of the Na,K-ATPase - low and high concentrations of H2O2 change the oxidative state of thiolate (S-) from Cys differently, resulting in the corresponding activation or inhibition of the enzyme. Targeting thiols of the Na,K-ATPase tunes the activity of the Na,K-ATPase to the cellular demands and sustains the enzyme activity at the "optimal" level.


Subject(s)
Hydrogen Peroxide , Sodium-Potassium-Exchanging ATPase , Sodium-Potassium-Exchanging ATPase/chemistry , Reactive Oxygen Species/metabolism , Hydrogen Peroxide/metabolism , Oxidation-Reduction , Cysteine
18.
Protein Expr Purif ; 200: 106167, 2022 12.
Article in English | MEDLINE | ID: mdl-36057422

ABSTRACT

The ß1-subunit of the Na+/K+-ATPase is a cell membrane protein, beyond its classic functions, it is also a cell adhesion molecule. ß1-subunits on the lateral membrane of dog kidney epithelial cells trans-interact with ß1-subunits from another neighboring cells. The ß-ß interaction is essential for the formation and stabilization of intercellular junctions. Previous studies on site-directed mutagenesis and in silico revealed that the interaction interface involves residues 198-207 and 221-229. However, it is necessary to report the interaction interface at the structural level experimentally. Here, we describe the successful cloning, overexpression in E. coli, and purification of the extracellular domain of the ß1-subunit from inclusion bodies. Experimental characterization by size exclusion chromatography and DLS indicated similar hydrodynamic properties of the protein refolded. Structural analysis by circular dichroism and Raman spectroscopy revealed the secondary structures in the folded protein of type ß-sheet, α-helix, random coil, and turn. We also performed ß1-ß1 interaction assays with the recombinant protein, showing dimers' formation (6xHisß1-ß1). Given our results, the recombinant extracellular domain of the ß1-subunit is highly similar to the native protein, therefore the current work in our laboratory aims to characterize at the atomic level the interaction interface between EDß1.


Subject(s)
Escherichia coli , Sodium-Potassium-Exchanging ATPase , Animals , Cell Adhesion Molecules/metabolism , Cell Membrane/metabolism , Dogs , Epithelial Cells , Escherichia coli/genetics , Escherichia coli/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism
19.
Am J Hum Genet ; 102(3): 505-514, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29499166

ABSTRACT

Although mutations in more than 90 genes are known to cause CMT, the underlying genetic cause of CMT remains unknown in more than 50% of affected individuals. The discovery of additional genes that harbor CMT2-causing mutations increasingly depends on sharing sequence data on a global level. In this way-by combining data from seven countries on four continents-we were able to define mutations in ATP1A1, which encodes the alpha1 subunit of the Na+,K+-ATPase, as a cause of autosomal-dominant CMT2. Seven missense changes were identified that segregated within individual pedigrees: c.143T>G (p.Leu48Arg), c.1775T>C (p.Ile592Thr), c.1789G>A (p.Ala597Thr), c.1801_1802delinsTT (p.Asp601Phe), c.1798C>G (p.Pro600Ala), c.1798C>A (p.Pro600Thr), and c.2432A>C (p.Asp811Ala). Immunostaining peripheral nerve axons localized ATP1A1 to the axolemma of myelinated sensory and motor axons and to Schmidt-Lanterman incisures of myelin sheaths. Two-electrode voltage clamp measurements on Xenopus oocytes demonstrated significant reduction in Na+ current activity in some, but not all, ouabain-insensitive ATP1A1 mutants, suggesting a loss-of-function defect of the Na+,K+ pump. Five mutants fall into a remarkably narrow motif within the helical linker region that couples the nucleotide-binding and phosphorylation domains. These findings identify a CMT pathway and a potential target for therapy development in degenerative diseases of peripheral nerve axons.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Genes, Dominant , Mutation/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Child , Family , Female , Humans , Male , Middle Aged , Pedigree , Sodium-Potassium-Exchanging ATPase/chemistry , Young Adult
20.
Article in English | MEDLINE | ID: mdl-33476762

ABSTRACT

Na+/K+-ATPase (NKA) function is inhibited by Bufadienolides (BD), a group of cardiotonic steroids (CTS) primarily produced by anurans of the Bufonidae family, such as Rhinella marina. This study characterized the presence of α and ß NKA subunit isoforms in R. marina via RNAseq in four tissues: oocytes, skin, heart, and skeletal muscle. Transcripts encoding three α-like isoforms (α1, α2, α3) and three ß-like isoforms (ß1, ß2, ß4) were identified. The amino acid sequence of α1-like isoform shared 99.4% identity with the α1 isoform previously published for R. marina. Sequences for α2, α3, and ß4 from R. marina were previously unavailable. The first extracellular loop in the α2-like isoform in R. marina showed similar substitutions to those found in their susceptible homologues in other taxa (L/Q111T and S119T); in contrast, this same loop in α3-like isoform showed similar substitutions (Q111L and G120R) to those reported for toad-eating animals such as snakes, which suggests relatively lower affinity for CTS. Docking results showed that all three α-like isoforms identified in R. marina transcriptomes have low affinity to CTS compared to the susceptible α1 isoform of Sus scrofa (pig), with α1-like isoform being the most resistant. The tissue-specific RNAseq results showed the following expression of NKA α-like and ß-like subunit isoforms: Oocytes expressed α1 and ß1; skin α1, ß1, and low levels of ß2; heart α1, α3, and ß1; skeletal muscle α1, ß4, with low levels of α2, α3, and ß1. R. marina could be used as an important model for future structural, functional and pharmacological studies of NKA and its isoforms.


Subject(s)
Base Sequence , Bufanolides/chemistry , Bufo marinus/metabolism , Sodium-Potassium-Exchanging ATPase/chemistry , Animals , Bufonidae , Hydrogen Bonding , Molecular Docking Simulation , Muscle, Skeletal/metabolism , Oocytes/cytology , Oocytes/metabolism , Phylogeny , Principal Component Analysis , Protein Isoforms , Ranidae , Sodium-Potassium-Exchanging ATPase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL