1
|
Chkadua G, Nozadze E, Tsakadze L, Shioshvili L, Arutinova N, Leladze M, Dzneladze S, Javakhishvili M, Jariashvili T, Petriashvili E. The effect of cytochrome c on Na,K-ATPase. J Bioenerg Biomembr 2024; 56:221-234. [PMID: 38517564 DOI: 10.1007/s10863-024-10012-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 03/09/2024] [Indexed: 03/24/2024]
Abstract
Na,K-ATPase is a crucial enzyme responsible for maintaining Na+, K+-gradients across the cell membrane, which is essential for numerous physiological processes within various organs and tissues. Due to its significance in cellular physiology, inhibiting Na,K-ATPase can have profound physiological consequences. This characteristic makes it a target for various pharmacological applications, and drugs that modulate the pump's activity are thus used in the treatment of several medical conditions. Cytochrome c (Cytc) is a protein with dual functions in the cell. In the mitochondria, it is essential for ATP synthesis and energy production. However, in response to apoptotic stimuli, it is released into the cytosol, where it triggers programmed cell death through the intrinsic apoptosis pathway. Aside from its role in canonical intrinsic apoptosis, Cytc also plays additional roles. For instance, Cytc participates in certain non-apoptotic functions -those which are less well-understood in comparison to its role in apoptosis. Within this in vitro study, we have shown the impact of Cytc on Na,K-ATPase for the first time. Cytc has a biphasic action on Na,K-ATPase, with activation at low concentrations (0.06 ng/ml; 6 ng/ml) and inhibition at high concentration (120 ng/ml). Cytc moreover displays isoform/subunit specificity and regulates the Na+ form of the enzyme, while having no effect on the activity or kinetic parameters of the K+-dependent form of the enzyme. Changing the affinity of p-chloromercuribenzoic acid (PCMB) by Cytc is therefore both a required and sufficient condition for confirming that PCMB and Cytc share the same target, namely the thiol groups of cysteine in Na,K-ATPase.
Collapse
Affiliation(s)
- Gvantsa Chkadua
- Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Str, 0160, Tbilisi, Georgia.
| | - Eka Nozadze
- Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Str, 0160, Tbilisi, Georgia
| | - Leila Tsakadze
- Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Str, 0160, Tbilisi, Georgia
| | - Lia Shioshvili
- Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Str, 0160, Tbilisi, Georgia
| | - Nana Arutinova
- Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Str, 0160, Tbilisi, Georgia
| | - Marine Leladze
- Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Str, 0160, Tbilisi, Georgia
| | - Sopio Dzneladze
- Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Str, 0160, Tbilisi, Georgia
| | - Maia Javakhishvili
- Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Str, 0160, Tbilisi, Georgia
| | | | | |
Collapse
|
2
|
Contreras RG, Torres-Carrillo A, Flores-Maldonado C, Shoshani L, Ponce A. Na +/K +-ATPase: More than an Electrogenic Pump. Int J Mol Sci 2024; 25:6122. [PMID: 38892309 PMCID: PMC11172918 DOI: 10.3390/ijms25116122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
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 β-subunits as cell adhesion molecules in glia and epithelial cells.
Collapse
Affiliation(s)
| | | | | | | | - Arturo Ponce
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, Mexico City 07360, Mexico; (R.G.C.); (A.T.-C.); (C.F.-M.); (L.S.)
| |
Collapse
|
3
|
Blaustein MP, Hamlyn JM. Sensational site: the sodium pump ouabain-binding site and its ligands. Am J Physiol Cell Physiol 2024; 326:C1120-C1177. [PMID: 38223926 PMCID: PMC11193536 DOI: 10.1152/ajpcell.00273.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/22/2023] [Accepted: 01/10/2024] [Indexed: 01/16/2024]
Abstract
Cardiotonic steroids (CTS), used by certain insects, toads, and rats for protection from predators, became, thanks to Withering's trailblazing 1785 monograph, the mainstay of heart failure (HF) therapy. In the 1950s and 1960s, we learned that the CTS receptor was part of the sodium pump (NKA) and that the Na+/Ca2+ exchanger was critical for the acute cardiotonic effect of digoxin- and ouabain-related CTS. This "settled" view was upended by seven revolutionary observations. First, subnanomolar ouabain sometimes stimulates NKA while higher concentrations are invariably inhibitory. Second, endogenous ouabain (EO) was discovered in the human circulation. Third, in the DIG clinical trial, digoxin only marginally improved outcomes in patients with HF. Fourth, cloning of NKA in 1985 revealed multiple NKA α and β subunit isoforms that, in the rodent, differ in their sensitivities to CTS. Fifth, the NKA is a cation pump and a hormone receptor/signal transducer. EO binding to NKA activates, in a ligand- and cell-specific manner, several protein kinase and Ca2+-dependent signaling cascades that have widespread physiological effects and can contribute to hypertension and HF pathogenesis. Sixth, all CTS are not equivalent, e.g., ouabain induces hypertension in rodents while digoxin is antihypertensinogenic ("biased signaling"). Seventh, most common rodent hypertension models require a highly ouabain-sensitive α2 NKA and the elevated blood pressure is alleviated by EO immunoneutralization. These numerous phenomena are enabled by NKA's intricate structure. We have just begun to understand the endocrine role of the endogenous ligands and the broad impact of the ouabain-binding site on physiology and pathophysiology.
Collapse
Affiliation(s)
- Mordecai P Blaustein
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - John M Hamlyn
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
4
|
Palmgren M. P-type ATPases: Many more enigmas left to solve. J Biol Chem 2023; 299:105352. [PMID: 37838176 PMCID: PMC10654040 DOI: 10.1016/j.jbc.2023.105352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/16/2023] Open
Abstract
P-type ATPases constitute a large ancient super-family of primary active pumps that have diverse substrate specificities ranging from H+ to phospholipids. The significance of these enzymes in biology cannot be overstated. They are structurally related, and their catalytic cycles alternate between high- and low-affinity conformations that are induced by phosphorylation and dephosphorylation of a conserved aspartate residue. In the year 1988, all P-type sequences available by then were analyzed and five major families, P1 to P5, were identified. Since then, a large body of knowledge has accumulated concerning the structure, function, and physiological roles of members of these families, but only one additional family, P6 ATPases, has been identified. However, much is still left to be learned. For each family a few remaining enigmas are presented, with the intention that they will stimulate interest in continued research in the field. The review is by no way comprehensive and merely presents personal views with a focus on evolution.
Collapse
Affiliation(s)
- Michael Palmgren
- Department of Plant and Environmental Sciences, University of Copenhagen, Frederiksberg C, Denmark.
| |
Collapse
|
5
|
Palmgren M. Evolution of the sodium pump. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119511. [PMID: 37301269 DOI: 10.1016/j.bbamcr.2023.119511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/16/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023]
Abstract
Eukaryotic plasma membranes (PMs) are energized by electrogenic P-type ATPases that generate either Na+ or H+ motive forces to drive Na+ and H+ dependent transport processes, respectively. For this purpose, animal rely on Na+/K+-ATPases whereas fungi and plants employ PM H+-ATPases. Prokaryotes, on the other hand, depend on H+ or Na+-motive electron transport complexes to energize their cell membranes. This raises the question as to why and when electrogenic Na+ and H+ pumps evolved? Here it is shown that prokaryotic Na+/K+-ATPases have near perfect conservation of binding sites involved in coordination of three Na+ and two K+ ions. Such pumps are rare in Eubacteria but are common in methanogenic Archaea where they often are found together with P-type putative PM H+-ATPases. With some exceptions, Na+/K+-ATPases and PM H+-ATPases are found everywhere in the eukaryotic tree of life, but never together in animals, fungi and land plants. It is hypothesized that Na+/K+-ATPases and PM H+-ATPases evolved in methanogenic Archaea to support the bioenergetics of these ancestral organisms, which can utilize both H+ and Na+ as energy currencies. Both pumps must have been simultaneously present in the first eukaryotic cell, but during diversification of the major eukaryotic kingdoms, and at the time animals diverged from fungi, animals kept Na+/K+-ATPases but lost PM H+-ATPases. At the same evolutionary branch point, fungi did loose Na+/K+-ATPases, and their role was taken over by PM H+-ATPases. An independent but similar scenery emerged during terrestrialization of plants: they lost Na+/K+-ATPases but kept PM H+-ATPases.
Collapse
Affiliation(s)
- Michael Palmgren
- Department of Plant and Environmental Sciences, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark.
| |
Collapse
|
6
|
Peluffo RD, Hernández JA. The Na +,K +-ATPase and its stoichiometric ratio: some thermodynamic speculations. Biophys Rev 2023; 15:539-552. [PMID: 37681108 PMCID: PMC10480117 DOI: 10.1007/s12551-023-01082-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/18/2023] [Indexed: 09/09/2023] Open
Abstract
Almost seventy years after its discovery, the sodium-potassium adenosine triphosphatase (the sodium pump) located in the cell plasma membrane remains a source of novel mechanistic and physiologic findings. A noteworthy feature of this enzyme/transporter is its robust stoichiometric ratio under physiological conditions: it sequentially counter-transports three sodium ions and two potassium ions against their electrochemical potential gradients per each hydrolyzed ATP molecule. Here we summarize some present knowledge about the sodium pump and its physiological roles, and speculate whether energetic constraints may have played a role in the evolutionary selection of its characteristic stoichiometric ratio.
Collapse
Affiliation(s)
- R. Daniel Peluffo
- Group of Biophysical Chemistry, Department of Biological Sciences, CENUR Litoral Norte, Universidad de La República, Rivera 1350, CP: 50000 Salto, Uruguay
| | - Julio A. Hernández
- Biophysics and Systems Biology Section, Department of Cell and Molecular Biology, Facultad de Ciencias, Universidad de La República, Iguá 4225, CP: 11400 Montevideo, Uruguay
| |
Collapse
|
7
|
Zhang X, Lee W, Bian JS. Recent Advances in the Study of Na +/K +-ATPase in Neurodegenerative Diseases. Cells 2022; 11:cells11244075. [PMID: 36552839 PMCID: PMC9777075 DOI: 10.3390/cells11244075] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Na+/K+-ATPase (NKA), a large transmembrane protein, is expressed in the plasma membrane of most eukaryotic cells. It maintains resting membrane potential, cell volume and secondary transcellular transport of other ions and neurotransmitters. NKA consumes about half of the ATP molecules in the brain, which makes NKA highly sensitive to energy deficiency. Neurodegenerative diseases (NDDs) are a group of diseases characterized by chronic, progressive and irreversible neuronal loss in specific brain areas. The pathogenesis of NDDs is sophisticated, involving protein misfolding and aggregation, mitochondrial dysfunction and oxidative stress. The protective effect of NKA against NDDs has been emerging gradually in the past few decades. Hence, understanding the role of NKA in NDDs is critical for elucidating the underlying pathophysiology of NDDs and identifying new therapeutic targets. The present review focuses on the recent progress involving different aspects of NKA in cellular homeostasis to present in-depth understanding of this unique protein. Moreover, the essential roles of NKA in NDDs are discussed to provide a platform and bright future for the improvement of clinical research in NDDs.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Weithye Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Jin-Song Bian
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
- Correspondence:
| |
Collapse
|
8
|
Blaustein MP, Gottlieb SS, Hamlyn JM, Leenen FHH. Whither digitalis? What we can still learn from cardiotonic steroids about heart failure and hypertension. Am J Physiol Heart Circ Physiol 2022; 323:H1281-H1295. [PMID: 36367691 DOI: 10.1152/ajpheart.00362.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Cloning of the "Na+ pump" (Na+,K+-ATPase or NKA) and identification of a circulating ligand, endogenous ouabain (EO), a cardiotonic steroid (CTS), triggered seminal discoveries regarding EO and its NKA receptor in cardiovascular function and the pathophysiology of heart failure (HF) and hypertension. Cardiotonic digitalis preparations were a preferred treatment for HF for two centuries, but digoxin was only marginally effective in a large clinical trial (1997). This led to diminished digoxin use. Missing from the trial, however, was any consideration that endogenous CTS might influence digitalis' efficacy. Digoxin, at therapeutic concentrations, acutely inhibits NKA but, remarkably, antagonizes ouabain's action. Prolonged treatment with ouabain, but not digoxin, causes hypertension in rodents; in this model, digoxin lowers blood pressure (BP). Furthermore, NKA-bound ouabain and digoxin modulate different protein kinase signaling pathways and have disparate long-term cardiovascular effects. Reports of "brain ouabain" led to the elucidation of a new, slow neuromodulatory pathway in the brain; locally generated EO and the α2 NKA isoform help regulate sympathetic drive to the heart and vasculature. The roles of EO and α2 NKA have been studied by EO assay, ouabain-resistant mutation of α2 NKA, and immunoneutralization of EO with ouabain-binding Fab fragments. The NKA α2 CTS binding site and its endogenous ligand are required for BP elevation in many common hypertension models and full expression of cardiac remodeling and dysfunction following pressure overload or myocardial infarction. Understanding how endogenous CTS impact hypertension and HF pathophysiology and therapy should foster reconsideration of digoxin's therapeutic utility.
Collapse
Affiliation(s)
- Mordecai P Blaustein
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Stephen S Gottlieb
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - John M Hamlyn
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Frans H H Leenen
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| |
Collapse
|
9
|
Multipurpose Na + ions mediate excitation and cellular homeostasis: Evolution of the concept of Na + pumps and Na +/Ca 2+ exchangers. Cell Calcium 2020; 87:102166. [PMID: 32006802 DOI: 10.1016/j.ceca.2020.102166] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/14/2022]
Abstract
Ionic signalling is the most ancient form of regulation of cellular functions in response to environmental challenges. Signals, mediated by Na+ fluxes and spatio-temporal fluctuations of Na+ concentration in cellular organelles and cellular compartments contribute to the most fundamental cellular processes such as membrane excitability and energy production. At the very core of ionic signalling lies the Na+-K+ ATP-driven pump (or NKA) which creates trans-plasmalemmal ion gradients that sustain ionic fluxes through ion channels and numerous Na+-dependent transporters that maintain cellular and tissue homeostasis. Here we present a brief account of the history of research into NKA, Na+ -dependent transporters and Na+ signalling.
Collapse
|
10
|
Zhao C, Yu Y, Zhang Y, Shen J, Jiang L, Sheng G, Zhang W, Xu L, Jiang K, Mao S, Jiang P, Gao F. β-Catenin Controls the Electrophysiologic Properties of Skeletal Muscle Cells by Regulating the α2 Isoform of Na +/K +-ATPase. Front Neurosci 2019; 13:831. [PMID: 31440132 PMCID: PMC6693565 DOI: 10.3389/fnins.2019.00831] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 07/25/2019] [Indexed: 12/20/2022] Open
Abstract
β-Catenin is a key component of the canonical Wnt signaling pathway. It has been shown to have an important role in formation of the neuromuscular junction. Our previous studies showed that in the absence of β-catenin, the resting membrane potential (RMP) is depolarized in muscle cells and expression of the α2 subunit of sodium/potassium adenosine triphosphatase (α2NKA) is reduced. To understand the underlying mechanisms, we investigated the electrophysiologic properties of a primary cell line derived from mouse myoblasts (C2C12 cells) that were transfected with small-interfering RNAs and over-expressed plasmids targeting β-catenin. We found that the RMP was depolarized in β-catenin knocked-down C2C12 cells and was unchanged in β-catenin over-expressed muscle cells. An action potential (AP) was not released by knockdown or over-expression of β-catenin. α2NKA expression was reduced by β-catenin knockdown, and increased by β-catenin over-expression. We showed that β-catenin could interact physically with α2NKA (but not with α1NKA) in muscle cells. NKA activity and α2NKA content in the cell membranes of skeletal muscle cells were modulated positively by β-catenin. These results suggested that β-catenin (at least in part) regulates the RMP and AP in muscle cells, and does so by regulating α2NKA.
Collapse
Affiliation(s)
- Congying Zhao
- Department of Neurology, Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yonglin Yu
- Department of Rehabilitation, Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Zhang
- Department of Neurology, Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jue Shen
- Department of Neurology, Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lihua Jiang
- Department of Neurology, Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Guoxia Sheng
- Department of Neurology, Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weiqin Zhang
- Department of Neurology, Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lu Xu
- Department of Neurology, Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kewen Jiang
- Department of Neurology, Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Biobank, Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shanshan Mao
- Department of Neurology, Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Scientific Research Office, Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Peifang Jiang
- Department of Neurology, Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Gao
- Department of Neurology, Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Nepal N, Arthur S, Sundaram U. Unique Regulation of Na-K-ATPase during Growth and Maturation of Intestinal Epithelial Cells. Cells 2019; 8:cells8060593. [PMID: 31208048 PMCID: PMC6628168 DOI: 10.3390/cells8060593] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 06/09/2019] [Accepted: 06/13/2019] [Indexed: 12/25/2022] Open
Abstract
Na-K-ATPase on the basolateral membrane provides the favorable transcellular Na gradient for the proper functioning of Na-dependent nutrient co-transporters on the brush border membrane (BBM) of enterocytes. As cells mature from crypts to villus, Na-K-ATPase activity doubles, to accommodate for the increased BBM Na-dependent nutrient absorption. However, the mechanism of increased Na-K-ATPase activity during the maturation of enterocytes is not known. Therefore, this study aimed to determine the mechanisms involved in the functional transition of Na-K-ATPase during the maturation of crypts to villus cells. Na-K-ATPase activity gradually increased as IEC-18 cells matured in vitro from day 0 (crypts) through day 4 (villus) of post-confluence. mRNA abundance and Western blot studies showed no change in the levels of Na-K-ATPase subunits α1 and β1 from 0 to 4 days post-confluent cells. However, Na-K-ATPase α1 phosphorylation levels on serine and tyrosine, but not threonine, residues gradually increased. These data indicate that as enterocytes mature from crypt-like to villus-like in culture, the functional activity of Na-K-ATPase increases secondary to altered affinity of the α1 subunit to extracellular K+, in order to accommodate the functional preference of the intestinal cell type. This altered affinity is likely due to increased phosphorylation of the α1 subunit, specifically at serine and tyrosine residues.
Collapse
Affiliation(s)
- Niraj Nepal
- Department of Clinical and Translational Sciences and Appalachian Clinical and Translational Science Institute, Joan C. Edwards School of Medicine, Marshall University, 1600 Medical Center Drive, Huntington, WV 25701, USA.
| | - Subha Arthur
- Department of Clinical and Translational Sciences and Appalachian Clinical and Translational Science Institute, Joan C. Edwards School of Medicine, Marshall University, 1600 Medical Center Drive, Huntington, WV 25701, USA.
| | - Uma Sundaram
- Department of Clinical and Translational Sciences and Appalachian Clinical and Translational Science Institute, Joan C. Edwards School of Medicine, Marshall University, 1600 Medical Center Drive, Huntington, WV 25701, USA.
| |
Collapse
|
12
|
Li XC, Zheng X, Chen X, Zhao C, Zhu D, Zhang J, Zhuo JL. Genetic and genomic evidence for an important role of the Na +/H + exchanger 3 in blood pressure regulation and angiotensin II-induced hypertension. Physiol Genomics 2019; 51:97-108. [PMID: 30849009 PMCID: PMC6485378 DOI: 10.1152/physiolgenomics.00122.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The sodium (Na+)/hydrogen (H+) exchanger 3 (NHE3) and sodium-potassium adenosine triphosphatase (Na+/K+-ATPase) are two of the most important Na+ transporters in the proximal tubules of the kidney. On the apical membrane side, NHE3 primarily mediates the entry of Na+ into and the exit of H+ from the proximal tubules, directly and indirectly being responsible for reabsorbing ~50% of filtered Na+ in the proximal tubules of the kidney. On the basolateral membrane side, Na+/K+-ATPase serves as a powerful engine driving Na+ out of, while pumping K+ into the proximal tubules against their concentration gradients. While the roles of NHE3 and Na+/K+-ATPase in proximal tubular Na+ transport under in vitro conditions are well recognized, their respective contributions to the basal blood pressure regulation and angiotensin II (ANG II)-induced hypertension remain poorly understood. Recently, we have been fortunate to be able to use genetically modified mouse models with global, kidney- or proximal tubule-specific deletion of NHE3 to directly determine the cause and effect relationship between NHE3, basal blood pressure homeostasis, and ANG II-induced hypertension at the whole body, kidney and/or proximal tubule levels. The purpose of this article is to review the genetic and genomic evidence for an important role of NHE3 with a focus in the regulation of basal blood pressure and ANG II-induced hypertension, as we learned from studies using global, kidney- or proximal tubule-specific NHE3 knockout mice. We hypothesize that NHE3 in the proximal tubules is necessary for maintaining basal blood pressure homeostasis and the development of ANG II-induced hypertension.
Collapse
Affiliation(s)
- Xiao C Li
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Xiaowen Zheng
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Xu Chen
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Chunling Zhao
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Dongmin Zhu
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Jianfeng Zhang
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Jia L Zhuo
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| |
Collapse
|
13
|
Gonzalez-Vicente A, Saez F, Monzon CM, Asirwatham J, Garvin JL. Thick Ascending Limb Sodium Transport in the Pathogenesis of Hypertension. Physiol Rev 2019; 99:235-309. [PMID: 30354966 DOI: 10.1152/physrev.00055.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The thick ascending limb plays a key role in maintaining water and electrolyte balance. The importance of this segment in regulating blood pressure is evidenced by the effect of loop diuretics or local genetic defects on this parameter. Hormones and factors produced by thick ascending limbs have both autocrine and paracrine effects, which can extend prohypertensive signaling to other structures of the nephron. In this review, we discuss the role of the thick ascending limb in the development of hypertension, not as a sole participant, but one that works within the rich biological context of the renal medulla. We first provide an overview of the basic physiology of the segment and the anatomical considerations necessary to understand its relationship with other renal structures. We explore the physiopathological changes in thick ascending limbs occurring in both genetic and induced animal models of hypertension. We then discuss the racial differences and genetic defects that affect blood pressure in humans through changes in thick ascending limb transport rates. Throughout the text, we scrutinize methodologies and discuss the limitations of research techniques that, when overlooked, can lead investigators to make erroneous conclusions. Thus, in addition to advancing an understanding of the basic mechanisms of physiology, the ultimate goal of this work is to understand our research tools, to make better use of them, and to contextualize research data. Future advances in renal hypertension research will require not only collection of new experimental data, but also integration of our current knowledge.
Collapse
Affiliation(s)
| | - Fara Saez
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Casandra M Monzon
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Jessica Asirwatham
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Jeffrey L Garvin
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| |
Collapse
|
14
|
Blaustein MP. The pump, the exchanger, and the holy spirit: origins and 40-year evolution of ideas about the ouabain-Na + pump endocrine system. Am J Physiol Cell Physiol 2017; 314:C3-C26. [PMID: 28971835 DOI: 10.1152/ajpcell.00196.2017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Two prescient 1953 publications set the stage for the elucidation of a novel endocrine system: Schatzmann's report that cardiotonic steroids (CTSs) are all Na+ pump inhibitors, and Szent-Gyorgi's suggestion that there is an endogenous "missing screw" in heart failure that CTSs like digoxin may replace. In 1977 I postulated that an endogenous Na+ pump inhibitor acts as a natriuretic hormone and simultaneously elevates blood pressure (BP) in salt-dependent hypertension. This hypothesis was based on the idea that excess renal salt retention promoted the secretion of a CTS-like hormone that inhibits renal Na+ pumps and salt reabsorption. The hormone also inhibits arterial Na+ pumps, elevates myocyte Na+ and promotes Na/Ca exchanger-mediated Ca2+ gain. This enhances vasoconstriction and arterial tone-the hallmark of hypertension. Here I describe how those ideas led to the discovery that the CTS-like hormone is endogenous ouabain (EO), a key factor in the pathogenesis of hypertension and heart failure. Seminal observations that underlie the still-emerging picture of the EO-Na+ pump endocrine system in the physiology and pathophysiology of multiple organ systems are summarized. Milestones include: 1) cloning the Na+ pump isoforms and physiological studies of mutated pumps in mice; 2) discovery that Na+ pumps are also EO-triggered signaling molecules; 3) demonstration that ouabain, but not digoxin, is hypertensinogenic; 4) elucidation of EO's roles in kidney development and cardiovascular and renal physiology and pathophysiology; 5) discovery of "brain ouabain", a component of a novel hypothalamic neuromodulatory pathway; and 6) finding that EO and its brain receptors modulate behavior and learning.
Collapse
Affiliation(s)
- Mordecai P Blaustein
- Departments of Physiology and Medicine, University of Maryland School of Medicine , Baltimore, Maryland
| |
Collapse
|
15
|
Blaustein MP. How does pressure overload cause cardiac hypertrophy and dysfunction? High-ouabain affinity cardiac Na + pumps are crucial. Am J Physiol Heart Circ Physiol 2017; 313:H919-H930. [PMID: 28733446 PMCID: PMC5792198 DOI: 10.1152/ajpheart.00131.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/17/2022]
Abstract
Left ventricular hypertrophy is frequently observed in hypertensive patients and is believed to be due to the pressure overload and cardiomyocyte stretch. Three recent reports on mice with genetically engineered Na+ pumps, however, have demonstrated that cardiac ouabain-sensitive α2-Na+ pumps play a key role in the pathogenesis of transaortic constriction-induced hypertrophy. Hypertrophy was delayed/attenuated in mice with mutant, ouabain-resistant α2-Na+ pumps and in mice with cardiac-selective knockout or transgenic overexpression of α2-Na+ pumps. The latter, seemingly paradoxical, findings can be explained by comparing the numbers of available (ouabain-free) high-affinity (α2) ouabain-binding sites in wild-type, knockout, and transgenic hearts. Conversely, hypertrophy was accelerated in α2-ouabain-resistant (R) mice in which the normally ouabain-resistant α1-Na+ pumps were mutated to an ouabain-sensitive (S) form (α1S/Sα2R/R or "SWAP" vs. wild-type or α1R/R α2S/S mice). Furthermore, transaortic constriction-induced hypertrophy in SWAP mice was prevented/reversed by immunoneutralizing circulating endogenous ouabain (EO). These findings show that EO and its receptor, ouabain-sensitive α2, are critical factors in pressure overload-induced cardiac hypertrophy. This complements reports linking elevated plasma EO to hypertension, cardiac hypertrophy, and failure in humans and elucidates the underappreciated role of the EO-Na+ pump pathway in cardiovascular disease.
Collapse
Affiliation(s)
- Mordecai P. Blaustein
- Departments of Physiology and Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
16
|
Cui X, Xie Z. Protein Interaction and Na/K-ATPase-Mediated Signal Transduction. Molecules 2017; 22:molecules22060990. [PMID: 28613263 PMCID: PMC6152704 DOI: 10.3390/molecules22060990] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/01/2017] [Accepted: 06/02/2017] [Indexed: 02/05/2023] Open
Abstract
The Na/K-ATPase (NKA), or Na pump, is a member of the P-type ATPase superfamily. In addition to pumping ions across cell membrane, it is engaged in assembly of multiple protein complexes in the plasma membrane. This assembly allows NKA to perform many non-pumping functions including signal transduction that are important for animal physiology and disease progression. This article will focus on the role of protein interaction in NKA-mediated signal transduction, and its potential utility as target for developing new therapeutics.
Collapse
Affiliation(s)
- Xiaoyu Cui
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA.
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA.
| |
Collapse
|
17
|
Subcellular localization of Na/K-ATPase isoforms in ventricular myocytes. J Mol Cell Cardiol 2017; 108:158-169. [PMID: 28587810 DOI: 10.1016/j.yjmcc.2017.05.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 05/29/2017] [Accepted: 05/31/2017] [Indexed: 11/22/2022]
Abstract
The sodium/potassium ATPase (NKA) is essential for establishing the normal intracellular [Na+] and [K+] and transmembrane gradients that are essential for many cellular functions, including cardiac electrophysiology and contractility. Different NKA isoforms exhibit differential expression levels, cellular localization, and function in different tissues and species. Prior work has indicated that the NKA-α1 isoform is quantitatively predominant in cardiac myocytes, but that the α2 isoform is preferentially concentrated in the transverse tubules (TT), possibly at junctions with the sarcoplasmic reticulum (SR) where α2 may preferentially modulate cardiac contractility. Here we measured subcellular localization of NKA-α1 and α2 using super-resolution microscopy (STED and STORM) and isoform-selective antibodies in mouse ventricular myocytes. We confirm the preferential localization of NKA-α2 in TT vs. surface sarcolemma, but also show that α2 is relatively excluded from longitudinal TT elements. In contrast NKA-α1 is relatively uniformly expressed in all three sarcolemmal regions. We also tested the hypothesis that NKA-α2 (vs. α1) is preferentially concentrated at SR junctional sites near ryanodine receptors (RyR2). The results refute this hypothesis, in that NKA-α1 and α2 were equally close to RyR2 at the TT, with no preferential NKA isoform localization near RyR2. We conclude that in contrast to relatively uniform NKA-α1 distribution, NKA-α2 is preferentially concentrated in the truly transverse (and not longitudinal) TT elements. However, NKA-α2 does not preferentially cluster at RyR2 junctions, so the TT NKA-α2 concentration may suffice for preferential effects of NKA-α2 inhibition on cardiac contractility.
Collapse
|
18
|
Tejral G, Sopko B, Necas A, Schoner W, Amler E. Computer modelling reveals new conformers of the ATP binding loop of Na +/K +-ATPase involved in the transphosphorylation process of the sodium pump. PeerJ 2017; 5:e3087. [PMID: 28316890 PMCID: PMC5354106 DOI: 10.7717/peerj.3087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 02/14/2017] [Indexed: 01/02/2023] Open
Abstract
Hydrolysis of ATP by Na+/K+-ATPase, a P-Type ATPase, catalyzing active Na+ and K+ transport through cellular membranes leads transiently to a phosphorylation of its catalytical α-subunit. Surprisingly, three-dimensional molecular structure analysis of P-type ATPases reveals that binding of ATP to the N-domain connected by a hinge to the P-domain is much too far away from the Asp369 to allow the transfer of ATP’s terminal phosphate to its aspartyl-phosphorylation site. In order to get information for how the transfer of the γ-phosphate group of ATP to the Asp369 is achieved, analogous molecular modeling of the M4–M5 loop of ATPase was performed using the crystal data of Na+/K+-ATPase of different species. Analogous molecular modeling of the cytoplasmic loop between Thr338 and Ile760 of the α2-subunit of Na+/K+-ATPase and the analysis of distances between the ATP binding site and phosphorylation site revealed the existence of two ATP binding sites in the open conformation; the first one close to Phe475 in the N-domain, the other one close to Asp369 in the P-domain. However, binding of Mg2+•ATP to any of these sites in the “open conformation” may not lead to phosphorylation of Asp369. Additional conformations of the cytoplasmic loop were found wobbling between “open conformation” <==> “semi-open conformation <==> “closed conformation” in the absence of 2Mg2+•ATP. The cytoplasmic loop’s conformational change to the “semi-open conformation”—characterized by a hydrogen bond between Arg543 and Asp611—triggers by binding of 2Mg2+•ATP to a single ATP site and conversion to the “closed conformation” the phosphorylation of Asp369 in the P-domain, and hence the start of Na+/K+-activated ATP hydrolysis.
Collapse
Affiliation(s)
- Gracian Tejral
- Department of Biophysics, 2nd Faculty of Medicine, Charles University Prague, Prague, Czech Republic; Laboratory of Tissue Engineering, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Bruno Sopko
- Department of Medical Chemistry and Clinical Biochemistry, 2nd Faculty of Medicine, Charles University Prague , Prague , Czech Republic
| | - Alois Necas
- Small Animal Clinic, Faculty of Veterinary Medicine, University of Veterinary and Pharmaceutical Science , Brno , Czech Republic
| | - Wilhelm Schoner
- Institute of Biochemistry and Endocrinology, University of Giessen , Giessen , Germany
| | - Evzen Amler
- Department of Biophysics, 2nd Faculty of Medicine, Charles University Prague, Prague, Czech Republic; Laboratory of Tissue Engineering, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| |
Collapse
|
19
|
Blaustein MP, Chen L, Hamlyn JM, Leenen FHH, Lingrel JB, Wier WG, Zhang J. Pivotal role of α2 Na + pumps and their high affinity ouabain binding site in cardiovascular health and disease. J Physiol 2016; 594:6079-6103. [PMID: 27350568 DOI: 10.1113/jp272419] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/18/2016] [Indexed: 12/13/2022] Open
Abstract
Reduced smooth muscle (SM)-specific α2 Na+ pump expression elevates basal blood pressure (BP) and increases BP sensitivity to angiotensin II (Ang II) and dietary NaCl, whilst SM-α2 overexpression lowers basal BP and decreases Ang II/salt sensitivity. Prolonged ouabain infusion induces hypertension in rodents, and ouabain-resistant mutation of the α2 ouabain binding site (α2R/R mice) confers resistance to several forms of hypertension. Pressure overload-induced heart hypertrophy and failure are attenuated in cardio-specific α2 knockout, cardio-specific α2 overexpression and α2R/R mice. We propose a unifying hypothesis that reconciles these apparently disparate findings: brain mechanisms, activated by Ang II and high NaCl, regulate sympathetic drive and a novel neurohumoral pathway mediated by both brain and circulating endogenous ouabain (EO). Circulating EO modulates ouabain-sensitive α2 Na+ pump activity and Ca2+ transporter expression and, via Na+ /Ca2+ exchange, Ca2+ homeostasis. This regulates sensitivity to sympathetic activity, Ca2+ signalling and arterial and cardiac contraction.
Collapse
Affiliation(s)
- Mordecai P Blaustein
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| | - Ling Chen
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - John M Hamlyn
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Frans H H Leenen
- Hypertension Unit, University of Ottawa Heart Institute, Ottawa, ON, Canada, K1Y 4W7
| | - Jerry B Lingrel
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0524, USA
| | - W Gil Wier
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jin Zhang
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| |
Collapse
|
20
|
Kinoshita PF, Leite JA, Orellana AMM, Vasconcelos AR, Quintas LEM, Kawamoto EM, Scavone C. The Influence of Na(+), K(+)-ATPase on Glutamate Signaling in Neurodegenerative Diseases and Senescence. Front Physiol 2016; 7:195. [PMID: 27313535 PMCID: PMC4890531 DOI: 10.3389/fphys.2016.00195] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/17/2016] [Indexed: 12/17/2022] Open
Abstract
Decreased Na(+), K(+)-ATPase (NKA) activity causes energy deficiency, which is commonly observed in neurodegenerative diseases. The NKA is constituted of three subunits: α, β, and γ, with four distinct isoforms of the catalytic α subunit (α1-4). Genetic mutations in the ATP1A2 gene and ATP1A3 gene, encoding the α2 and α3 subunit isoforms, respectively can cause distinct neurological disorders, concurrent to impaired NKA activity. Within the central nervous system (CNS), the α2 isoform is expressed mostly in glial cells and the α3 isoform is neuron-specific. Mutations in ATP1A2 gene can result in familial hemiplegic migraine (FHM2), while mutations in the ATP1A3 gene can cause Rapid-onset dystonia-Parkinsonism (RDP) and alternating hemiplegia of childhood (AHC), as well as the cerebellar ataxia, areflexia, pescavus, optic atrophy and sensorineural hearing loss (CAPOS) syndrome. Data indicates that the central glutamatergic system is affected by mutations in the α2 isoform, however further investigations are required to establish a connection to mutations in the α3 isoform, especially given the diagnostic confusion and overlap with glutamate transporter disease. The age-related decline in brain α2∕3 activity may arise from changes in the cyclic guanosine monophosphate (cGMP) and cGMP-dependent protein kinase (PKG) pathway. Glutamate, through nitric oxide synthase (NOS), cGMP and PKG, stimulates brain α2∕3 activity, with the glutamatergic N-methyl-D-aspartate (NMDA) receptor cascade able to drive an adaptive, neuroprotective response to inflammatory and challenging stimuli, including amyloid-β. Here we review the NKA, both as an ion pump as well as a receptor that interacts with NMDA, including the role of NKA subunits mutations. Failure of the NKA-associated adaptive response mechanisms may render neurons more susceptible to degeneration over the course of aging.
Collapse
Affiliation(s)
- Paula F. Kinoshita
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Jacqueline A. Leite
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Ana Maria M. Orellana
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Andrea R. Vasconcelos
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Luis E. M. Quintas
- Laboratory of Biochemical and Molecular Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Elisa M. Kawamoto
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Cristoforo Scavone
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| |
Collapse
|
21
|
Feraille E, Dizin E. Coordinated Control of ENaC and Na+,K+-ATPase in Renal Collecting Duct. J Am Soc Nephrol 2016; 27:2554-63. [PMID: 27188842 DOI: 10.1681/asn.2016020124] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Tubular reabsorption of filtered sodium is tightly controlled to maintain body volume homeostasis. The rate of sodium transport by collecting duct (CD) cells varies widely in response to dietary sodium intake, GFR, circulating hormones, neural signals, and local regulatory factors. Reabsorption of filtered sodium by CD cells occurs via a two-step process. First, luminal sodium crosses the apical plasma membrane along its electrochemical gradient through epithelial sodium channels (ENaC). Intracellular sodium is then actively extruded into the interstitial space by the Na(+),K(+)-ATPase located along the basolateral membrane. Mismatch between sodium entry and exit induces variations in sodium intracellular concentration and cell volume that must be maintained within narrow ranges for control of vital cell functions. Therefore, renal epithelial cells display highly coordinated apical and basolateral sodium transport rates. We review evidence from experiments conducted in vivo and in cultured cells that indicates aldosterone and vasopressin, the two major hormones regulating sodium reabsorption by CD, generate a coordinated stimulation of apical ENaC and basolateral Na(+),K(+)-ATPase. Moreover, we discuss evidence suggesting that variations in sodium entry per se induce a coordinated change in Na(+),K(+)-ATPase activity through the signaling of protein kinases such as protein kinase A and p38 mitogen-activated protein kinase.
Collapse
Affiliation(s)
- Eric Feraille
- Department of Cell Biology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Eva Dizin
- Department of Cell Biology and Metabolism, University of Geneva, Geneva, Switzerland
| |
Collapse
|
22
|
Thabet R, Rouault JD, Ayadi H, Leignel V. Structural analysis of the α subunit of Na(+)/K(+) ATPase genes in invertebrates. Comp Biochem Physiol B Biochem Mol Biol 2016; 196-197:11-18. [PMID: 26812300 DOI: 10.1016/j.cbpb.2016.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 01/20/2016] [Accepted: 01/21/2016] [Indexed: 01/18/2023]
Abstract
The Na(+)/K(+) ATPase is a ubiquitous pump coordinating the transport of Na(+) and K(+) across the membrane of cells and its role is fundamental to cellular functions. It is heteromer in eukaryotes including two or three subunits (α, β and γ which is specific to the vertebrates). The catalytic functions of the enzyme have been attributed to the α subunit. Several complete α protein sequences are available, but only few gene structures were characterized. We identified the genomic sequences coding the α-subunit of the Na(+)/K(+) ATPase, from the whole-genome shotgun contigs (WGS), NCBI Genomes (chromosome), Genomic Survey Sequences (GSS) and High Throughput Genomic Sequences (HTGS) databases across distinct phyla. One copy of the α subunit gene was found in Annelida, Arthropoda, Cnidaria, Echinodermata, Hemichordata, Mollusca, Placozoa, Porifera, Platyhelminthes, Urochordata, but the nematodes seem to possess 2 to 4 copies. The number of introns varied from 0 (Platyhelminthes) to 26 (Porifera); and their localization and length are also highly variable. Molecular phylogenies (Maximum Likelihood and Maximum Parsimony methods) showed some clusters constituted by (Chordata/(Echinodermata/Hemichordata)) or (Plathelminthes/(Annelida/Mollusca)) and a basal position for Porifera. These structural analyses increase our knowledge about the evolutionary events of the α subunit genes in the invertebrates.
Collapse
Affiliation(s)
- Rahma Thabet
- University of Sfax, Laboratory of Biodiversity and Aquatic Ecosystems UR/11ES72, Ecology and Planktonology, Department of Life Sciences, Road Soukra Km 3.5, BP1171, 3000, Sfax, Tunisia
| | - J-D Rouault
- Laboratoire Evolution, Genomes et Speciation, UPR9034, CNRS, 91198 Gif-sur-Yvette, France
| | - Habib Ayadi
- University of Sfax, Laboratory of Biodiversity and Aquatic Ecosystems UR/11ES72, Ecology and Planktonology, Department of Life Sciences, Road Soukra Km 3.5, BP1171, 3000, Sfax, Tunisia
| | - Vincent Leignel
- Université du Maine, Laboratoire Mer Molecule Sante EA 2160 FR-CNRS 3473 IUML, 72085 Le Mans, France.
| |
Collapse
|
23
|
High CO2 Leads to Na,K-ATPase Endocytosis via c-Jun Amino-Terminal Kinase-Induced LMO7b Phosphorylation. Mol Cell Biol 2015; 35:3962-73. [PMID: 26370512 DOI: 10.1128/mcb.00813-15] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 09/09/2015] [Indexed: 01/04/2023] Open
Abstract
The c-Jun amino-terminal kinase (JNK) plays a role in inflammation, proliferation, apoptosis, and cell adhesion and cell migration by phosphorylating paxillin and β-catenin. JNK phosphorylation downstream of AMP-activated protein kinase (AMPK) activation is required for high CO2 (hypercapnia)-induced Na,K-ATPase endocytosis in alveolar epithelial cells. Here, we provide evidence that during hypercapnia, JNK promotes the phosphorylation of LMO7b, a scaffolding protein, in vitro and in intact cells. LMO7b phosphorylation was blocked by exposing the cells to the JNK inhibitor SP600125 and by infecting cells with dominant-negative JNK or AMPK adenovirus. The knockdown of the endogenous LMO7b or overexpression of mutated LMO7b with alanine substitutions of five potential JNK phosphorylation sites (LMO7b-5SA) or only Ser-1295 rescued both LMO7b phosphorylation and the hypercapnia-induced Na,K-ATPase endocytosis. Moreover, high CO2 promoted the colocalization and interaction of LMO7b and the Na,K-ATPase α1 subunit at the plasma membrane, which were prevented by SP600125 or by transfecting cells with LMO7b-5SA. Collectively, our data suggest that hypercapnia leads to JNK-induced LMO7b phosphorylation at Ser-1295, which facilitates the interaction of LMO7b with Na,K-ATPase at the plasma membrane promoting the endocytosis of Na,K-ATPase in alveolar epithelial cells.
Collapse
|
24
|
Su Y, Al-Lamki RS, Blake-Palmer KG, Best A, Golder ZJ, Zhou A, Karet Frankl FE. Physical and functional links between anion exchanger-1 and sodium pump. J Am Soc Nephrol 2014; 26:400-9. [PMID: 25012180 DOI: 10.1681/asn.2013101063] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Anion exchanger-1 (AE1) mediates chloride-bicarbonate exchange across the plasma membranes of erythrocytes and, via a slightly shorter transcript, kidney epithelial cells. On an omnivorous human diet, kidney AE1 is mainly active basolaterally in α-intercalated cells of the collecting duct, where it is functionally coupled with apical proton pumps to maintain normal acid-base homeostasis. The C-terminal tail of AE1 has an important role in its polarized membrane residency. We have identified the β1 subunit of Na(+),K(+)-ATPase (sodium pump) as a binding partner for AE1 in the human kidney. Kidney AE1 and β1 colocalized in renal α-intercalated cells and coimmunoprecipitated (together with the catalytic α1 subunit of the sodium pump) from human kidney membrane fractions. ELISA and fluorescence titration assays confirmed that AE1 and β1 interact directly, with a Kd value of 0.81 μM. GST-AE1 pull-down assays using human kidney membrane proteins showed that the last 11 residues of AE1 are important for β1 binding. siRNA-induced knockdown of β1 in cell culture resulted in a significant reduction in kidney AE1 levels at the cell membrane, whereas overexpression of kidney AE1 increased cell surface sodium pump levels. Notably, membrane staining of β1 was reduced throughout collecting ducts of AE1-null mouse kidney, where increased fractional excretion of sodium has been reported. These data suggest a requirement of β1 for proper kidney AE1 membrane residency, and that activities of AE1 and the sodium pump are coregulated in kidney.
Collapse
Affiliation(s)
- Ya Su
- Departments of Medical Genetics and
| | - Rafia S Al-Lamki
- Division of Renal Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | | | | | | | - Fiona E Karet Frankl
- Departments of Medical Genetics and Division of Renal Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
25
|
Zhao C, Zhang J, Li K, Yang J, Yu H, Duan S, Jiang K, Li X. β-Catenin regulates membrane potential in muscle cells by regulating the α2 subunit of Na,K-ATPase. Eur J Neurosci 2014; 40:2216-24. [PMID: 24674304 DOI: 10.1111/ejn.12564] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 02/05/2014] [Accepted: 02/18/2014] [Indexed: 01/28/2023]
Abstract
Muscle β-catenin has been shown to play a role in the formation of the neuromuscular junction (NMJ). Our previous studies showed that muscle-specific conditional knockout of β-catenin (HSA-β-cat(-/-) ) results in early postnatal death in mice. To understand the underlying mechanisms, we investigated the electrophysiological properties of muscle cells from HSA-β-cat(-/-) and control mice, and found that, in the absence of muscle β-catenin, the resting membrane potential (RMP) depolarised in muscle cells from the diaphragm, gastrocnemius and extensor digitorum longus muscles. Furthermore, in a primary line of mouse myoblasts (C2C12 cells) transfected with small-interfering RNAs targeting β-catenin, the RMP was depolarised as well. Finally, the expression levels of the α2 subunit of sodium/potassium adenosine triphosphatase were reduced by β-catenin knockdown in vitro or deletion in vivo. These results suggest a possible mechanism underlying the depolarised RMP in the absence of muscle β-catenin, and provide additional evidence supporting a role for β-catenin in the development of NMJs.
Collapse
Affiliation(s)
- Congying Zhao
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Xie JX, Li X, Xie Z. Regulation of renal function and structure by the signaling Na/K-ATPase. IUBMB Life 2013; 65:991-8. [PMID: 24323927 PMCID: PMC5375025 DOI: 10.1002/iub.1229] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 11/08/2013] [Indexed: 12/23/2022]
Abstract
The Na/K-ATPase as an essential ion pump was discovered more than 50 years ago (Skou (1989) Biochim. Biophys. Acta 1000, 439-446; Feraille and Doucet (2001) Physiol. Rev. 81, 345-418). The signaling function of Na/K-ATPase has been gradually appreciated over the last 20 years, first from the studies of regulatory effects of ouabain on cardiac cell growth. Several reviews on this topic have been written during the last few years (Schoner and Scheiner-Bobis (2007) Am. J. Physiol. Cell. Physiol. 293, C509-C536; Xie and Cai (2003) Mol. Interv. 3, 157 - 168; Bagrov et al. (2009) Pharmacol. Rev. 61, 9-38; Tian and Xie (2008) Physiology 23, 205-211; Fontana et al. (2013) FEBS J. 280, 5450-5455; Blanco and Wallace (2013) Am. J. Physiol. Renal Physiol. 305, F797-F812). This article will focus on the molecular mechanism of Na/K-ATPase-mediated signal transduction and its potential regulatory role in renal physiology and diseases.
Collapse
Affiliation(s)
- Jeffrey X Xie
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, USA
| | | | | |
Collapse
|
27
|
Holzinger F, Wink M. Mediation of cardiac glycoside insensitivity in the monarch butterfly (Danaus plexippus): Role of an amino acid substitution in the ouabain binding site of Na(+),K (+)-ATPase. J Chem Ecol 2013; 22:1921-37. [PMID: 24227116 DOI: 10.1007/bf02028512] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/1996] [Accepted: 05/16/1996] [Indexed: 12/01/2022]
Abstract
The Monarch butterfly (Danaus plexippus) sequesters cardiac glycosides (CG) for its chemical defense against predators. Larvae and adults of this butterfly are insensitive towards dietary cardiac glycosides, whereas other Lepidoptera are sensitive and intoxicated by ouabain. Ouabain inhibits Na(+),K(+)-ATPase by binding to its α-subunit. We have amplified and cloned the DNA-sequence encoding the respective ouabain binding site. Instead of the amino acid asparagine at position 122 in ouabain-sensitive insects, the Monarch has a histidine in the putative ouabain binding site, which consists of 12 amino acids. Starting with the CG-sensitive Na(+),K(+)-ATPase gene fromDrosophila, we converted pos. 122 to a histidine residue as inDanaus plexippus by site-directed mutagenesis. Human embryonic kidney cells (HEK) (which are sensitive to ouabain) were transfected with the mutated Na(+),K(+)-ATPase gene in a pSVDF-expression vector and showed a transient expression of the mutatedDrosophila Na(+),K(+)-ATPase. When treated with ouabain, the transfected cells tolerated ouabain at a concentration of 50 mM, whereas untransformed controls or controls transfected with the unmutatedDrosophila gene, showed a substantial mortality. This result implies that the asparagine to histidine exchange contributes to ouabain insensitivity in the Monarch. In two other CG-sequestering insects, e.g.,Danaus gilippus andSyntomeida epilais, the pattern of amino acid substitution differed, indicating that the Monarch has acquired this mutation independently during evolution.
Collapse
Affiliation(s)
- F Holzinger
- Institut für Pharmazeutische Biologie, Universität Heidelberg, Im Neuenheimer Feld 364, D-69120, Heidelberg, Germany
| | | |
Collapse
|
28
|
Abstract
The kidney plays a fundamental role in maintaining body salt and fluid balance and blood pressure homeostasis through the actions of its proximal and distal tubular segments of nephrons. However, proximal tubules are well recognized to exert a more prominent role than distal counterparts. Proximal tubules are responsible for reabsorbing approximately 65% of filtered load and most, if not all, of filtered amino acids, glucose, solutes, and low molecular weight proteins. Proximal tubules also play a key role in regulating acid-base balance by reabsorbing approximately 80% of filtered bicarbonate. The purpose of this review article is to provide a comprehensive overview of new insights and perspectives into current understanding of proximal tubules of nephrons, with an emphasis on the ultrastructure, molecular biology, cellular and integrative physiology, and the underlying signaling transduction mechanisms. The review is divided into three closely related sections. The first section focuses on the classification of nephrons and recent perspectives on the potential role of nephron numbers in human health and diseases. The second section reviews recent research on the structural and biochemical basis of proximal tubular function. The final section provides a comprehensive overview of new insights and perspectives in the physiological regulation of proximal tubular transport by vasoactive hormones. In the latter section, attention is particularly paid to new insights and perspectives learnt from recent cloning of transporters, development of transgenic animals with knockout or knockin of a particular gene of interest, and mapping of signaling pathways using microarrays and/or physiological proteomic approaches.
Collapse
Affiliation(s)
- Jia L Zhuo
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA.
| | | |
Collapse
|
29
|
Affiliation(s)
- R W Albers
- Laboratory of Neurochemistry, NINCDS, National Institutes of Health, Bethesda, MD 20892, U.S.A
| |
Collapse
|
30
|
Mercury toxicity on sodium pump and organoseleniums intervention: a paradox. J Biomed Biotechnol 2012; 2012:924549. [PMID: 22927724 PMCID: PMC3425867 DOI: 10.1155/2012/924549] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 07/01/2012] [Indexed: 12/21/2022] Open
Abstract
Mercury is an environmental poison, and the damage to living system is generally severe. The severity of mercury poisoning is consequent from the fact that it targets the thiol-containing enzymes, irreversibly oxidizing their critical thiol groups, consequently leading to an inactivation of the enzyme. The Na+/K+-ATPase is a sulfhydryl protein that is sensitive to Hg2+ assault. On the other hand, organoseleniums are a class of pharmacologically promising compounds with potent antioxidant effects. While Hg2+ oxidizes sulfhydryl groups of Na+/K+-ATPase under in vitro and in vivo conditions, the organoselenium compounds inhibit Na+/K+-ATPase in vitro but enhance its activities under in vivo conditions with concomitant increase in the level of endogenous thiols. Paradoxically, it appears that these two thiol oxidants can be used to counteract one another under in vivo conditions, and this hypothesis serves as the basis for this paper.
Collapse
|
31
|
Na(+), K(+)-ATPase subunit composition in a human chondrocyte cell line; evidence for the presence of α1, α3, β1, β2 and β3 isoforms. Int J Mol Sci 2012; 13:5019-5034. [PMID: 22606027 PMCID: PMC3344263 DOI: 10.3390/ijms13045019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 04/06/2012] [Accepted: 04/12/2012] [Indexed: 11/16/2022] Open
Abstract
Membrane transport systems participate in fundamental activities such as cell cycle control, proliferation, survival, volume regulation, pH maintenance and regulation of extracellular matrix synthesis. Multiple isoforms of Na(+), K(+)-ATPase are expressed in primary chondrocytes. Some of these isoforms have previously been reported to be expressed exclusively in electrically excitable cells (i.e., cardiomyocytes and neurons). Studying the distribution of Na(+), K(+)-ATPase isoforms in chondrocytes makes it possible to document the diversity of isozyme pairing and to clarify issues concerning Na(+), K(+)-ATPase isoform abundance and the physiological relevance of their expression. In this study, we investigated the expression of Na(+), K(+)-ATPase in a human chondrocyte cell line (C-20/A4) using a combination of immunological and biochemical techniques. A panel of well-characterized antibodies revealed abundant expression of the α1, β1 and β2 isoforms. Western blot analysis of plasma membranes confirmed the above findings. Na(+), K(+)-ATPase consists of multiple isozyme variants that endow chondrocytes with additional homeostatic control capabilities. In terms of Na(+), K(+)-ATPase expression, the C-20/A4 cell line is phenotypically similar to primary and in situ chondrocytes. However, unlike freshly isolated chondrocytes, C-20/A4 cells are an easily accessible and convenient in vitro model for the study of Na(+), K(+)-ATPase expression and regulation in chondrocytes.
Collapse
|
32
|
Weihrauch D, Donini A, O'Donnell MJ. Ammonia transport by terrestrial and aquatic insects. JOURNAL OF INSECT PHYSIOLOGY 2012; 58:473-87. [PMID: 22100291 DOI: 10.1016/j.jinsphys.2011.11.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 11/03/2011] [Accepted: 11/04/2011] [Indexed: 05/13/2023]
Abstract
Ammonia, an end product from amino acid and nucleic acid metabolism, is highly toxic for most animals. This review will provide an update on nitrogen metabolism in terrestrial and aquatic insects with emphasis on ammonia generation and transport. Aspects that will be discussed include metabolic pathways of nitrogenous compounds, the origin of ammonia and other nitrogenous waste products, ammonia toxicity, putative ammonia transporters as well as ammonia transport processes known in insects. Ammonia transport mechanisms in the mosquito Aedes aegypti, the tobacco hornworm Manduca sexta and the locust Schistocerca gregaria will be discussed in detail while providing additional, novel data.
Collapse
Affiliation(s)
- Dirk Weihrauch
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, Canada R3T2N2.
| | | | | |
Collapse
|
33
|
Gupta SP. Quantitative structure-activity relationship studies on Na+,K(+)-ATPase inhibitors. Chem Rev 2012; 112:3171-92. [PMID: 22360614 DOI: 10.1021/cr200097p] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Satya P Gupta
- Department of Applied Sciences, Meerut Institute of Engineering and Technology, Meerut-250 005, India.
| |
Collapse
|
34
|
Bucking C, Wood CM. Digestion of a single meal affects gene expression of ion and ammonia transporters and glutamine synthetase activity in the gastrointestinal tract of freshwater rainbow trout. J Comp Physiol B 2011; 182:341-50. [PMID: 21994022 DOI: 10.1007/s00360-011-0622-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 09/23/2011] [Accepted: 09/27/2011] [Indexed: 12/25/2022]
Abstract
Experiments on freshwater rainbow trout, Oncorhynchus mykiss, demonstrated how digestion affected the transcriptional expression of gastrointestinal transporters following a single satiating meal (~3% body mass ration) after a 1-week fast. Quantitative real-time polymerase chain reaction was employed to measure the relative mRNA expression of three previously cloned and sequenced transporters [H(+)-K(+)-ATPase (HKA), Na(+)/HCO(3)(-) cotransporter (NBC), and the Rhesus glycoprotein (Rhbg1; an ammonia transporter)] over a 24-h time course following feeding. Plasma total ammonia increased about threefold from pre-feeding levels to 288 μmol l(-1), whereas total ammonia levels in chyme supernatant reached a sixfold higher value (1.8 mmol l(-1)) than plasma levels. Feeding did not appear to have a statistically significant effect on the relative mRNA expression of the gastric HKA or Rhbg1. However, the relative mRNA expression of gastric NBC was increased 24 h following the ingestion of a meal. Along the intestinal tract, feeding increased the relative mRNA expression of Rhbg1, but had no effect on the expression of NBC. Expression of the gastric HKA was undetectable in the intestinal tract of freshwater rainbow trout. Digestion increased the activity of glutamine synthetase in the posterior intestine at 12 and 24 h following feeding. This study is among the first to show that there are digestion-associated changes in gene expression and enzyme activity in the gastrointestinal tract of teleost fish illustrating the dynamic plasticity of this organ. These post-prandial changes occur over the relative short-term duration of digesting a single meal.
Collapse
Affiliation(s)
- Carol Bucking
- McMaster University, 1280 Main Street, West Hamilton, ON, L8S 4K1, Canada.
| | | |
Collapse
|
35
|
Christoph K, Beck FX, Neuhofer W. Osmoadaptation of Mammalian cells - an orchestrated network of protective genes. Curr Genomics 2011; 8:209-18. [PMID: 18645598 DOI: 10.2174/138920207781386979] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Revised: 01/27/2007] [Accepted: 03/03/2007] [Indexed: 11/22/2022] Open
Abstract
In mammals, the cells of the renal medulla are physiologically exposed to interstitial osmolalities several-fold higher that found in any other tissue. Nevertheless, these cells not only have the ability to survive in this harsh environment, but also to function normally, which is critical for maintenance of systemic electrolyte and fluid homeostasis. Over the last two decades, a substantial body of evidence has accumulated, indicating that sequential and well orchestrated genomic responses are required to provide tolerance to osmotic stress. This includes the enhanced expression and action of immediate-early genes, growth arrest and DNA damage inducible genes (GADDs), genes involved in cell cycle control and apoptosis, heat shock proteins, and ultimately that of genes involved in the intracellular accumulation of nonperturbing organic osmolytes. The present review summarizes the sequence of genomic responses conferring resistance against osmotic stress. In addition, the regulatory mechanisms mediating the coordinated genomic response to osmotic stress will be highlighted.
Collapse
Affiliation(s)
- Küper Christoph
- Department of Physiology, University of Munich, Munich, Germany
| | | | | |
Collapse
|
36
|
Yang Y, Dai M, Wilson TM, Omelchenko I, Klimek JE, Wilmarth PA, David LL, Nuttall AL, Gillespie PG, Shi X. Na+/K+-ATPase α1 identified as an abundant protein in the blood-labyrinth barrier that plays an essential role in the barrier integrity. PLoS One 2011; 6:e16547. [PMID: 21304972 PMCID: PMC3031570 DOI: 10.1371/journal.pone.0016547] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Accepted: 12/21/2010] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The endothelial-blood/tissue barrier is critical for maintaining tissue homeostasis. The ear harbors a unique endothelial-blood/tissue barrier which we term "blood-labyrinth-barrier". This barrier is critical for maintaining inner ear homeostasis. Disruption of the blood-labyrinth-barrier is closely associated with a number of hearing disorders. Many proteins of the blood-brain-barrier and blood-retinal-barrier have been identified, leading to significant advances in understanding their tissue specific functions. In contrast, capillaries in the ear are small in volume and anatomically complex. This presents a challenge for protein analysis studies, which has resulted in limited knowledge of the molecular and functional components of the blood-labyrinth-barrier. In this study, we developed a novel method for isolation of the stria vascularis capillary from CBA/CaJ mouse cochlea and provided the first database of protein components in the blood-labyrinth barrier as well as evidence that the interaction of Na(+)/K(+)-ATPase α1 (ATP1A1) with protein kinase C eta (PKCη) and occludin is one of the mechanisms of loud sound-induced vascular permeability increase. METHODOLOGY/PRINCIPAL FINDINGS Using a mass-spectrometry, shotgun-proteomics approach combined with a novel "sandwich-dissociation" method, more than 600 proteins from isolated stria vascularis capillaries were identified from adult CBA/CaJ mouse cochlea. The ion transporter ATP1A1 was the most abundant protein in the blood-labyrinth barrier. Pharmacological inhibition of ATP1A1 activity resulted in hyperphosphorylation of tight junction proteins such as occludin which increased the blood-labyrinth-barrier permeability. PKCη directly interacted with ATP1A1 and was an essential mediator of ATP1A1-initiated occludin phosphorylation. Moreover, this identified signaling pathway was involved in the breakdown of the blood-labyrinth-barrier resulting from loud sound trauma. CONCLUSIONS/SIGNIFICANCE The results presented here provide a novel method for capillary isolation from the inner ear and the first database on protein components in the blood-labyrinth-barrier. Additionally, we found that ATP1A1 interaction with PKCη and occludin was involved in the integrity of the blood-labyrinth-barrier.
Collapse
Affiliation(s)
- Yue Yang
- Department of Otolaryngology/Head and Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Min Dai
- Department of Otolaryngology/Head and Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Teresa M. Wilson
- Department of Otolaryngology/Head and Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Irina Omelchenko
- Department of Otolaryngology/Head and Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - John E. Klimek
- Proteomic Shared Resources, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Phillip A. Wilmarth
- Proteomic Shared Resources, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Larry L. David
- Proteomic Shared Resources, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Alfred L. Nuttall
- Department of Otolaryngology/Head and Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
- Kresge Hearing Research Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Otolaryngology, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Peter G. Gillespie
- Department of Otolaryngology/Head and Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
- Vollum Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Xiaorui Shi
- Department of Otolaryngology/Head and Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Otolaryngology, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
- The Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- * E-mail:
| |
Collapse
|
37
|
Rajasekaran SA, Huynh TP, Wolle DG, Espineda CE, Inge LJ, Skay A, Lassman C, Nicholas SB, Harper JF, Reeves AE, Ahmed MM, Leatherman JM, Mullin JM, Rajasekaran AK. Na,K-ATPase subunits as markers for epithelial-mesenchymal transition in cancer and fibrosis. Mol Cancer Ther 2010; 9:1515-24. [PMID: 20501797 DOI: 10.1158/1535-7163.mct-09-0832] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Epithelial-to-mesenchymal transition (EMT) is an important developmental process, participates in tissue repair, and occurs during pathologic processes of tumor invasiveness, metastasis, and tissue fibrosis. The molecular mechanisms leading to EMT are poorly understood. Although it is well documented that transforming growth factor (TGF)-beta plays a central role in the induction of EMT, the targets of TGF-beta signaling are poorly defined. We have shown earlier that Na,K-ATPase beta(1)-subunit levels are highly reduced in poorly differentiated kidney carcinoma cells in culture and in patients' tumor samples. In this study, we provide evidence that Na,K-ATPase is a new target of TGF-beta(1)-mediated EMT in renal epithelial cells, a model system used in studies of both cancer progression and fibrosis. We show that following treatment with TGF-beta(1), the surface expression of the beta(1)-subunit of Na,K-ATPase is reduced, before well-characterized EMT markers, and is associated with the acquisition of a mesenchymal phenotype. RNAi-mediated knockdown confirmed the specific involvement of the Na,K-ATPase beta(1)-subunit in the loss of the epithelial phenotype and exogenous overexpression of the Na,K-ATPase beta(1)-subunit attenuated TGF-beta(1)-mediated EMT. We further show that both Na,K-ATPase alpha- and beta-subunit levels are highly reduced in renal fibrotic tissues. These findings reveal for the first time that Na,K-ATPase is a target of TGF-beta(1)-mediated EMT and is associated with the progression of EMT in cancer and fibrosis.
Collapse
Affiliation(s)
- Sigrid A Rajasekaran
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware 19803, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kathirvel K, Parija SC. Role of Na-K ATPase enzyme in vascular response of goat ruminal artery. Indian J Pharmacol 2010; 41:68-71. [PMID: 20336220 PMCID: PMC2841235 DOI: 10.4103/0253-7613.51343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2008] [Revised: 12/20/2008] [Accepted: 04/01/2009] [Indexed: 11/04/2022] Open
Abstract
Objective: To study the role of Na+, K+- ATPase enzyme in the vascular response of goat ruminal artery. Materials and Methods: Ruminal artery was obtained in chilled aerated modified Krebs-Henseleit solution (KHS) from a local slaughterhouse and transported in ice for further processing. The endothelium intact arterial ring was mounted in a thermostatically controlled (37 ± 0.5°C) organ bath containing 20 ml of modified KHS (pH 7.4) bubbled with oxygen (95%) and CO2 (5%) under 2g tension. An equilibration of 90 min was allowed before addition of drugs into the bath. The responses were recorded isometrically in an automatic organ bath connected to PowerLab data acquisition system. In order to examine intact functional endothelium, ACh (10 μM) was added on the 5-HT (1.0 μM) - induced sustained contractile response. Similarly, functional characterization of Na+, K+-ATPase activity was done by K+-induced relaxation (10 μM-10 mM) in the absence and presence of ouabain (0.1 μM/ 0.1 mM), digoxin (0.1 μM) and barium (30 μM). Results: ACh (10−5 M) did not produce any relaxing effect on 5-HT-induced sustained contractile response suggesting that vascular endothelium has no significant influence on the activation of sodium pump by extracellular K+ in ruminal artery. Low concentration of Ba2+ (30 μM) (IC50: 0.479 mM) inhibited K+-induced relaxation suggesting Kir (inward rectifier) channel in part had role in K+-induced vasodilatation in ruminal artery. Vasorelaxant effect of KCl (10 μM-10 mM) in K+-free medium is also blocked by ouabain (0.1 μM and 0.1 mM) (IC50:0.398 mM and IC35: 1.36 mM), but not by digoxin (0.1 μM) (IC50 0.234 mM) suggesting that ouabain sensitive Na+, K+-ATPase isoform is present in the ruminal artery. Conclusion: In the goat ruminal artery functional regulation of sodium pump is partly mediated by K+ channel and ouabain sensitive Na+, K+ ATPase.
Collapse
Affiliation(s)
- K Kathirvel
- Department of Pharmacology and Toxicology, Faculty of Veterinary Sciences and Animal Husbandry, Orissa University of Agriculture and Technology, Bhubaneswar - 751 003, India
| | | |
Collapse
|
39
|
Rigoard P, Lapierre F. Rappels sur le nerf périphérique. Neurochirurgie 2009; 55:360-74. [DOI: 10.1016/j.neuchi.2009.08.156] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Accepted: 08/13/2009] [Indexed: 12/20/2022]
|
40
|
Helmstetter C, Reix N, T'Flachebba M, Pope RK, Secor SM, Le Maho Y, Lignot JH. Functional Changes with Feeding in the Gastro-Intestinal Epithelia of the Burmese Python (Python molurus). Zoolog Sci 2009; 26:632-8. [DOI: 10.2108/zsj.26.632] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
41
|
Hatori Y, Lewis D, Toyoshima C, Inesi G. Reaction cycle of Thermotoga maritima copper ATPase and conformational characterization of catalytically deficient mutants. Biochemistry 2009; 48:4871-80. [PMID: 19364131 PMCID: PMC2756213 DOI: 10.1021/bi900338n] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
![]()
Copper transport ATPases sustain important roles in homeostasis of heavy metals and delivery of copper to metalloenzymes. The copper transport ATPase from Thermotoga maritima (CopA) provides a useful system for mechanistic studies, due to its heterologous expression and stability. Its sequence comprises 726 amino acids, including the N-terminal metal binding domain (NMBD), three catalytic domains (A, N, and P), and a copper transport domain formed by eight helices, including the transmembrane metal binding site (TMBS). We performed functional characterization and conformational analysis by proteolytic digestion of WT and mutated (NMBD deletion or mutation) T. maritima CopA, comparing it with Archaeoglobus fulgidus CopA and Ca2+ ATPase. A specific feature of T. maritima CopA is ATP utilization in the absence of copper, to form a low-turnover phosphoenzyme intermediate, with a conformation similar to that obtained by phosphorylation with Pi or phosphate analogues. On the other hand, formation of an activated state requires copper binding to both NMBD and TMBS, with consequent conformational changes involving the NMBD and A domain. Proteolytic digestion analysis demonstrates A domain movements similar to those of other P-type ATPases to place the conserved TGES motif in the optimal position for catalytic assistance. We also studied an H479Q mutation (analogous to one of human copper ATPase ATP7B in Wilson disease) that inhibits ATPase activity. We found that, in spite of the H479Q mutation within the nucleotide binding domain, the mutant still binds ATP, yielding a phosphorylation transition state conformation. However, covalent phosphoryl transfer is not completed, and no catalytic turnover is observed.
Collapse
Affiliation(s)
- Yuta Hatori
- California Pacific Medical Center Research Institute, San Francisco, California 94107, USA
| | | | | | | |
Collapse
|
42
|
Gamba G, Friedman PA. Thick ascending limb: the Na(+):K (+):2Cl (-) co-transporter, NKCC2, and the calcium-sensing receptor, CaSR. Pflugers Arch 2009; 458:61-76. [PMID: 18982348 PMCID: PMC3584568 DOI: 10.1007/s00424-008-0607-1] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Accepted: 10/21/2008] [Indexed: 01/12/2023]
Abstract
The thick ascending limb of Henle's loop is a nephron segment that is vital to the formation of dilute and concentrated urine. This ability is accomplished by a consortium of functionally coupled proteins consisting of the apical Na(+):K(+):2Cl(-) co-transporter, the K(+) channel, and basolateral Cl(-) channel that mediate electroneutral salt absorption. In thick ascending limbs, salt absorption is importantly regulated by the calcium-sensing receptor. Genetic or pharmacological disruption impairing the function of any of these proteins results in Bartter syndrome. The thick ascending limb is also an important site of Ca(2+) and Mg(2+) absorption. Calcium-sensing receptor activation inhibits cellular Ca(2+) absorption induced by parathyroid hormone, as well as passive paracellular Ca(2+) transport. The present review discusses these functions and their genetic and molecular regulation.
Collapse
Affiliation(s)
- Gerardo Gamba
- Molecular Physiology Unit, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, 14000 Mexico City, Mexico
| | - Peter A. Friedman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
43
|
Bagrov AY, Shapiro JI, Fedorova OV. Endogenous cardiotonic steroids: physiology, pharmacology, and novel therapeutic targets. Pharmacol Rev 2009; 61:9-38. [PMID: 19325075 PMCID: PMC2763610 DOI: 10.1124/pr.108.000711] [Citation(s) in RCA: 392] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Endogenous cardiotonic steroids (CTS), also called digitalis-like factors, have been postulated to play important roles in health and disease for nearly half a century. Recent discoveries, which include the specific identification of endogenous cardenolide (endogenous ouabain) and bufadienolide (marinobufagenin) CTS in humans along with the delineation of an alternative mechanism by which CTS can signal through the Na(+)/K(+)-ATPase, have increased the interest in this field substantially. Although CTS were first considered important in the regulation of renal sodium transport and arterial pressure, more recent work implicates these hormones in the regulation of cell growth, differentiation, apoptosis, and fibrosis, the modulation of immunity and of carbohydrate metabolism, and the control of various central nervous functions and even behavior. This review focuses on the physiological interactions between CTS and other regulatory systems that may be important in the pathophysiology of essential hypertension, preeclampsia, end-stage renal disease, congestive heart failure, and diabetes mellitus. Based on our increasing understanding of the regulation of CTS as well as the molecular mechanisms of these hormone increases, we also discuss potential therapeutic strategies.
Collapse
Affiliation(s)
- Alexei Y Bagrov
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, 5600 Nathan Shock Dr., Baltimore, MD 21224, USA.
| | | | | |
Collapse
|
44
|
Rigoard S, Wager M, Buffenoir K, Bauche S, Giot JP, Maixent JM, Rigoard P. Principaux mécanismes impliqués dans la transmission synaptique au sein de l’appareil neuromusculaire. Neurochirurgie 2009; 55 Suppl 1:S22-33. [DOI: 10.1016/j.neuchi.2008.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Accepted: 03/24/2008] [Indexed: 10/21/2022]
|
45
|
Lee DI, Klein MG, Zhu W, Xiao RP, Gerzanich V, Xu KY. Activation of (Na+ + K+)-ATPase modulates cardiac L-type Ca2+ channel function. Mol Pharmacol 2009; 75:774-81. [PMID: 19122004 DOI: 10.1124/mol.108.052597] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cellular Ca(2+) signaling underlies diverse vital biological processes, including muscle contractility, memory encoding, fertilization, cell survival, and cell death. Despite extensive studies, the fundamental control mechanisms that regulate intracellular Ca(2+) movement remain enigmatic. We have found recently that activation of the (Na(+)+K(+))-ATPase markedly potentiates intracellular Ca(2+) transients and contractility of rat heart cells. Little is known about the pathway responsible for the activation of the (Na(+)+K(+))-ATPase-initiated Ca(2+) signaling. Here, we demonstrate a novel mechanism in which activation of the (Na(+)+K(+))-ATPase is coupled to increased L-type Ca(2+) channel function through a signaling cascade involving Src and ERK1/2 but not well established regulators of the channel, such as adrenergic receptor system or activation of PKA or CaMKII. We have also identified Ser(1928), a phosphorylation site for the alpha1 subunit of the L-type Ca(2+) channel that may participate in the activation of the (Na(+)+K(+))-ATPase-mediated Ca(2+) signaling. The findings reported here uncover a novel molecular cross-talk between activation of the (Na(+)+K(+))-ATPase and L-type Ca(2+) channel and provide new insights into Ca(2+) signaling mechanisms for deeper understanding of the nature of cellular Ca(2+) handling in heart.
Collapse
Affiliation(s)
- Dong I Lee
- Department of Biochemistry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | |
Collapse
|
46
|
Keller-Wood M, von Reitzenstein M, McCartney J. Is the fetal lung a mineralocorticoid receptor target organ? Induction of cortisol-regulated genes in the ovine fetal lung, kidney and small intestine. Neonatology 2009; 95:47-60. [PMID: 18787337 PMCID: PMC2654587 DOI: 10.1159/000151755] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Accepted: 01/28/2008] [Indexed: 01/29/2023]
Abstract
BACKGROUND Lung, kidney and small intestine are involved in fetal volume regulation and amniotic fluid secretion and play a pivotal role in the transition from intrauterine to extrauterine life. OBJECTIVE This study was performed to determine the ontogeny of mineralocorticoid receptors (MR) and glucocorticoid receptors (GR), and of MR- and GR-regulated genes and proteins, serum and glucocorticoid-induced kinase (Sgk-1), epithelial sodium channel (ENaC alpha), and Na,K-ATPase alpha1. METHODS Lung, renal cortex and medulla, and small intestine were collected from fetuses at 80, 100, 120, 130 and 145 days' gestation and from day 1 and 7 neonatal lambs. Real-time PCR was performed to determine mRNA concentration for MR, GR, the 11 beta-hydroxysteroid dehydrogenases (11 beta-HSD1 and 2), Sgk-1, ENaC alpha, and Na,K-ATPase alpha1. Protein expression of ENaC alpha and Na,K-ATPase alpha1 in whole cell and membrane fractions was determined by immunoblotting. RESULTS Expression of corticosteroid-induced genes in renal cortex increases at term; in small intestine the induction occurs postnatally. In contrast, in lung expression of MR and GR mRNAs were greater at 100 days to term than postnatally and 11 beta-HSD1 peaked at 145 days; the corticosteroid-induced genes also increased prenatally: Sgk-1 and ENaC alpha increased by 120 days, peaking at 145 days, and Na,K-ATPase alpha1 was greatest at 130 days. CONCLUSIONS The expression of high levels of MR and 11 beta-HSD1 in preterm fetal lung suggest low endogenous fetal cortisol may exert actions at the high affinity MR in vivo, leading to increases in expression of sodium channels important in the regulation of lung liquid secretion and reabsorption.
Collapse
Affiliation(s)
- Maureen Keller-Wood
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32610, USA.
| | | | | |
Collapse
|
47
|
Xu KY. Dual activity of the H1-H2 domain of the (Na(+)+K+)-ATPase. Biochem Biophys Res Commun 2008; 377:469-473. [PMID: 18848919 DOI: 10.1016/j.bbrc.2008.09.137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2008] [Accepted: 09/30/2008] [Indexed: 11/17/2022]
Abstract
(Na(+)+K(+))-ATPase is a target receptor of digitalis (cardiac glycoside) drugs. It has been demonstrated that the H1-H2 domain of the alpha-subunit of the (Na(+)+K(+))-ATPase is one of the digitalis drug interaction sites of the enzyme. Despite the extensive studies of the inhibitory effect of digitalis on the (Na(+)+K(+))-ATPase, the functional property of the H1-H2 domain of the enzyme and its role in regulating enzyme activity is not completely understood. Here we report a surprise finding: instead of inhibiting the enzyme, binding of a specific monoclonal antibody SSA78 to the H1-H2 domain of the (Na(+)+K(+))-ATPase elevates the catalytic activity of the enzyme. In the presence of low concentration of ouabain, monoclonal antibody SSA78 significantly protects enzyme function against ouabain-induced inhibition. However, higher concentration of ouabain completely inactivates the (Na(+)+K(+))-ATPase even in the presence of SSA78. These results suggest that the H1-H2 domain of the (Na(+)+K(+))-ATPase is capable of regulating enzyme function in two distinct ways for both ouabain-sensitive and -resistant forms of the enzyme: it increases the activity of the (Na(+)+K(+))-ATPase during its interaction with an activator; it also participates in the mechanism of digitalis or ouabain-induced inhibition of the enzyme. Understanding the dual activity of the H1-H2 domain will help better understand the structure-function relationships of the (Na(+)+K(+))-ATPase and the biological processes mediated by the enzyme.
Collapse
Affiliation(s)
- Kai Y Xu
- Department of Surgery, Division of Cardiac Surgery, University of Maryland School of Medicine, MSTF-434E, 10 South Pine Street, Baltimore, MD 21201, USA.
| |
Collapse
|
48
|
Silva ECC, Masui DC, Furriel RPM, Mantelatto FLM, McNamara JC, Barrabin H, Leone FA, Scofano HM, Fontes CFL. Regulation by the exogenous polyamine spermidine of Na,K-ATPase activity from the gills of the euryhaline swimming crab Callinectes danae (Brachyura, Portunidae). Comp Biochem Physiol B Biochem Mol Biol 2008; 149:622-9. [PMID: 18272416 DOI: 10.1016/j.cbpb.2007.12.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2007] [Revised: 12/28/2007] [Accepted: 12/28/2007] [Indexed: 11/25/2022]
Abstract
Euryhaline crustaceans rarely hyporegulates and employ the driving force of the Na,K-ATPase, located at the basal surface of the gill epithelium, to maintain their hemolymph osmolality within a range compatible with cell function during hyper-regulation. Since polyamine levels increase during the adaptation of crustaceans to hyperosmotic media, we investigate the effect of exogenous polyamines on Na,K-ATPase activity in the posterior gills of Callinectes danae, a euryhaline swimming crab. Polyamine inhibition was dependent on cation concentration, charge and size in the following order: spermine>spermidine>putrescine. Spermidine affected K(0.5) values for Na(+) with minor alterations in K(0.5) values for K(+) and NH(4)(+), causing a decrease in maximal velocities under saturating Na(+), K(+) and NH(4)(+) concentrations. Phosphorylation measurements in the presence of 20 microM ATP revealed that the Na,K-ATPase possesses a high affinity site for this substrate. In the presence of 10 mM Na(+), both spermidine and spermine inhibited formation of the phosphoenzyme; however, in the presence of 100 mM Na(+), the addition of these polyamines allowed accumulation of the phosphoenzyme. The polyamines inhibited pumping activity, both by competing with Na(+) at the Na(+)-binding site, and by inhibiting enzyme dephosphorylation. These findings suggest that polyamine-induced inhibition of Na,K-ATPase activity may be physiologically relevant during migration to fully marine environments.
Collapse
Affiliation(s)
- E C C Silva
- Instituto de Bioquímica Médica, Laboratório de Estrutura e Regulação de Proteínas e ATPases, Programa de Biologia Estrutural, CCS, Bloco H, 2 andar, sala 26, 21941-590, RJ, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Characterization of NKIP: a novel, Na+/K+-ATPase interacting protein mediates neural differentiation and apoptosis. Exp Cell Res 2007; 314:463-77. [PMID: 18096156 DOI: 10.1016/j.yexcr.2007.11.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2007] [Revised: 11/05/2007] [Accepted: 11/08/2007] [Indexed: 01/17/2023]
Abstract
Cellular differentiation and programmed cell death are tightly controlled to maintain tissue homeostasis and proper organ function. In a screen for apoptosis specific gene products, we isolated an immediate early apoptosis response gene from myelomonocytic stem cells that appears to play a key regulatory role in a number of cell types and may be of particular importance in cells of the central nervous system. The gene's 28 kDa protein product interacts with the C-terminal ectodomain of the Na+/K+-ATPase (NKA) beta 1 subunit and was therefore named NKIP (NKA Interacting Protein). NKIP is coexpressed with NKA, localizes to lysosomes and the endoplasmic reticulum and is predominantly expressed in excitable tissues including polarized epithelia and the central nervous system. NKIP has been characterized as an endogenous suppressor of the NKA as reduction of NKIP in PC12 cells significantly increases NKA activity. In pluripotent NT2 progenitor cells, NKIP induced rapidly K+-level-dependent cell death. NKIP overexpression induced growth factor-independent neurite outgrowth, which was associated with MEK-independent phosphorylation of the transcription factor ERK1/2. Thus, we have identified NKIP as an important novel protein that interacts to the NKA complex, influencing cellular ion balance, induction of apoptosis and neuronal differentiation.
Collapse
|
50
|
Rajasekaran SA, Beyenbach KW, Rajasekaran AK. Interactions of tight junctions with membrane channels and transporters. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2007; 1778:757-69. [PMID: 18086552 DOI: 10.1016/j.bbamem.2007.11.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2007] [Revised: 11/02/2007] [Accepted: 11/09/2007] [Indexed: 12/22/2022]
Abstract
Tight junctions are unique organelles in epithelial cells. They are localized to the apico-lateral region and essential for the epithelial cell transport functions. The paracellular transport process that occurs via tight junctions is extensively studied and is intricately regulated by various extracellular and intracellular signals. Fine regulation of this transport pathway is crucial for normal epithelial cell functions. Among factors that control tight junction permeability are ions and their transporters. However, this area of research is still in its infancy and much more needs to be learned about how these molecules regulate tight junction structure and functions. In this review we have attempted to compile literature on ion transporters and channels involved in the regulation of tight junctions.
Collapse
Affiliation(s)
- Sigrid A Rajasekaran
- The Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|