1
|
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
|
2
|
Levic DS, Bagnat M. Polarized transport of membrane and secreted proteins during lumen morphogenesis. Semin Cell Dev Biol 2023; 133:65-73. [PMID: 35307284 PMCID: PMC9481742 DOI: 10.1016/j.semcdb.2022.03.016] [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: 12/03/2021] [Revised: 03/09/2022] [Accepted: 03/12/2022] [Indexed: 10/18/2022]
Abstract
A ubiquitous feature of animal development is the formation of fluid-filled cavities or lumina, which transport gases and fluids across tissues and organs. Among different species, lumina vary drastically in size, scale, and complexity. However, all lumen formation processes share key morphogenetic principles that underly their development. Fundamentally, a lumen simply consists of epithelial cells that encapsulate a continuous internal space, and a common way of building a lumen is via opening and enlarging by filling it with fluid and/or macromolecules. Here, we discuss how polarized targeting of membrane and secreted proteins regulates lumen formation, mainly focusing on ion transporters in vertebrate model systems. We also discuss mechanistic differences observed among invertebrates and vertebrates and describe how the unique properties of the Na+/K+-ATPase and junctional proteins can promote polarization of immature epithelia to build lumina de novo in developing organs.
Collapse
Affiliation(s)
- Daniel S Levic
- Department of Cell Biology, Duke University, Durham, NC 27710, USA.
| | - Michel Bagnat
- Department of Cell Biology, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
3
|
McCann C, Quinteros M, Adelugba I, Morgada MN, Castelblanco AR, Davis EJ, Lanzirotti A, Hainer SJ, Vila AJ, Navea JG, Padilla-Benavides T. The mitochondrial Cu+ transporter PiC2 (SLC25A3) is a target of MTF1 and contributes to the development of skeletal muscle in vitro. Front Mol Biosci 2022; 9:1037941. [PMID: 36438658 PMCID: PMC9682256 DOI: 10.3389/fmolb.2022.1037941] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022] Open
Abstract
The loading of copper (Cu) into cytochrome c oxidase (COX) in mitochondria is essential for energy production in cells. Extensive studies have been performed to characterize mitochondrial cuproenzymes that contribute to the metallation of COX, such as Sco1, Sco2, and Cox17. However, limited information is available on the upstream mechanism of Cu transport and delivery to mitochondria, especially through Cu-impermeable membranes, in mammalian cells. The mitochondrial phosphate transporter SLC25A3, also known as PiC2, binds Cu+ and transports the ion through these membranes in eukaryotic cells, ultimately aiding in the metallation of COX. We used the well-established differentiation model of primary myoblasts derived from mouse satellite cells, wherein Cu availability is necessary for growth and maturation, and showed that PiC2 is a target of MTF1, and its expression is both induced during myogenesis and favored by Cu supplementation. PiC2 deletion using CRISPR/Cas9 showed that the transporter is required for proliferation and differentiation of primary myoblasts, as both processes are delayed upon PiC2 knock-out. The effects of PiC2 deletion were rescued by the addition of Cu to the growth medium, implying the deleterious effects of PiC2 knockout in myoblasts may be in part due to a failure to deliver sufficient Cu to the mitochondria, which can be compensated by other mitochondrial cuproproteins. Co-localization and co-immunoprecipitation of PiC2 and COX also suggest that PiC2 may participate upstream in the copper delivery chain into COX, as verified by in vitro Cu+-transfer experiments. These data indicate an important role for PiC2 in both the delivery of Cu to the mitochondria and COX, favoring the differentiation of primary myoblasts.
Collapse
|
4
|
Roa-Velázquez D, Xoconostle-Cázares B, Benítez-Cardoza CG, Ortega-López J, Shoshani L, Morales-Ríos E, Gallardo-Hernández S. Expression, purification, and refolding of the recombinant extracellular domain β 1-subunit of the dog Na +/K +-ATPase of the epithelial cells. Protein Expr Purif 2022; 200:106167. [PMID: 36057422 DOI: 10.1016/j.pep.2022.106167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/25/2022]
Abstract
The β1-subunit of the Na+/K+-ATPase is a cell membrane protein, beyond its classic functions, it is also a cell adhesion molecule. β1-subunits on the lateral membrane of dog kidney epithelial cells trans-interact with β1-subunits from another neighboring cells. The β-β interaction is essential for the formation and stabilization of intercellular junctions. Previous studies on site-directed mutagenesis and in silico revealed that the interaction interface involves residues 198-207 and 221-229. However, it is necessary to report the interaction interface at the structural level experimentally. Here, we describe the successful cloning, overexpression in E. coli, and purification of the extracellular domain of the β1-subunit from inclusion bodies. Experimental characterization by size exclusion chromatography and DLS indicated similar hydrodynamic properties of the protein refolded. Structural analysis by circular dichroism and Raman spectroscopy revealed the secondary structures in the folded protein of type β-sheet, α-helix, random coil, and turn. We also performed β1-β1 interaction assays with the recombinant protein, showing dimers' formation (6xHisβ1-β1). Given our results, the recombinant extracellular domain of the β1-subunit is highly similar to the native protein, therefore the current work in our laboratory aims to characterize at the atomic level the interaction interface between EDβ1.
Collapse
Affiliation(s)
- Daniela Roa-Velázquez
- Programa de Doctorado en Nanociencias y Nanotecnología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, Ciudad de México, 07360, Mexico.
| | - Beatriz Xoconostle-Cázares
- Departamento de Bioingeniería y Biotecnología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, Ciudad de México, 07360, Mexico.
| | - Claudia G Benítez-Cardoza
- Laboratorio de Investigación Bioquímica, Escuela Nacional de Medicina y Homeopatía-Instituto Politécnico Nacional, Guillermo Massieu Helguera 239, Ciudad de México, 07320, Mexico.
| | - Jaime Ortega-López
- Departamento de Bioingeniería y Biotecnología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, Ciudad de México, 07360, Mexico.
| | - Liora Shoshani
- Departamento de Fisiología Biofísica y Neurociencias, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, Ciudad de México, 07360, Mexico.
| | - Edgar Morales-Ríos
- Departamento de Bioquímica, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, Ciudad de México, 07360, Mexico.
| | - Salvador Gallardo-Hernández
- Departamento de Física, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, Ciudad de México, 07360, Mexico.
| |
Collapse
|
5
|
Roldán ML, Ramírez-Salinas GL, Martinez-Archundia M, Cuellar-Perez F, Vilchis-Nestor CA, Cancino-Diaz JC, Shoshani L. The β2-Subunit (AMOG) of Human Na+, K+-ATPase Is a Homophilic Adhesion Molecule. Int J Mol Sci 2022; 23:ijms23147753. [PMID: 35887102 PMCID: PMC9322774 DOI: 10.3390/ijms23147753] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/05/2022] [Accepted: 07/05/2022] [Indexed: 12/10/2022] Open
Abstract
The β2 subunit of Na+, K+-ATPase was originally identified as the adhesion molecule on glia (AMOG) that mediates the adhesion of astrocytes to neurons in the central nervous system and that is implicated in the regulation of neurite outgrowth and neuronal migration. While β1 isoform have been shown to trans-interact in a species-specific mode with the β1 subunit on the epithelial neighboring cell, the β2 subunit has been shown to act as a recognition molecule on the glia. Nevertheless, none of the works have identified the binding partner of β2 or described its adhesion mechanism. Until now, the interactions pronounced for β2/AMOG are heterophilic cis-interactions. In the present report we designed experiments that would clarify whether β2 is a cell–cell homophilic adhesion molecule. For this purpose, we performed protein docking analysis, cell–cell aggregation, and protein–protein interaction assays. We observed that the glycosylated extracellular domain of β2/AMOG can make an energetically stable trans-interacting dimer. We show that CHO (Chinese Hamster Ovary) fibroblasts transfected with the human β2 subunit become more adhesive and make large aggregates. The treatment with Tunicamycin in vivo reduced cell aggregation, suggesting the participation of N-glycans in that process. Protein–protein interaction assay in vivo with MDCK (Madin-Darby canine kidney) or CHO cells expressing a recombinant β2 subunit show that the β2 subunits on the cell surface of the transfected cell lines interact with each other. Overall, our results suggest that the human β2 subunit can form trans-dimers between neighboring cells when expressed in non-astrocytic cells, such as fibroblasts (CHO) and epithelial cells (MDCK).
Collapse
Affiliation(s)
- María Luisa Roldán
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, 2508 IPN Ave., San Pedro Zacatenco, Ciudad de México 07360, Mexico; (M.L.R.); (F.C.-P.); (C.A.V.-N.)
| | - Gema Lizbeth Ramírez-Salinas
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Circuito, Mario de La Cueva S/N, C.U., Coyoacán, Ciudad de México 04510, Mexico;
| | - Marlet Martinez-Archundia
- Laboratorio de Modelado Molecular, Bioinformática y Diseño de Fármacos, Departamento de Posgrado Escuela Superior de Medicina del Instituto Politécnico Nacional, Salvador Díaz Mirón esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, Ciudad de México 11340, Mexico;
| | - Francisco Cuellar-Perez
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, 2508 IPN Ave., San Pedro Zacatenco, Ciudad de México 07360, Mexico; (M.L.R.); (F.C.-P.); (C.A.V.-N.)
| | - Claudia Andrea Vilchis-Nestor
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, 2508 IPN Ave., San Pedro Zacatenco, Ciudad de México 07360, Mexico; (M.L.R.); (F.C.-P.); (C.A.V.-N.)
| | - Juan Carlos Cancino-Diaz
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Salvador Díaz Mirón esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, Ciudad de México 11340, Mexico;
| | - Liora Shoshani
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, 2508 IPN Ave., San Pedro Zacatenco, Ciudad de México 07360, Mexico; (M.L.R.); (F.C.-P.); (C.A.V.-N.)
- Correspondence: ; Tel.: +52-55-5747-3360
| |
Collapse
|
6
|
Esmail S, Manolson MF. Advances in understanding N-glycosylation structure, function, and regulation in health and disease. Eur J Cell Biol 2021; 100:151186. [PMID: 34839178 DOI: 10.1016/j.ejcb.2021.151186] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/14/2021] [Accepted: 11/18/2021] [Indexed: 01/17/2023] Open
Abstract
N-linked glycosylation is a post-translational modification crucial for membrane protein folding, stability and other cellular functions. Alteration of membrane protein N-glycans is implicated in wide range of pathological conditions including cancer metastasis, chronic inflammatory diseases, and viral pathogenesis. Even though the roles of N-glycans have been studied extensively, our knowledge of their mechanisms remains unclear due to the lack of detailed structural analysis of the N-glycome. Mapping the N-glycome landscape will open new avenues to explore disease mechanisms and identify novel therapeutic targets. This review discusses the diverse structure of N-linked glycans, the function and regulation of N-glycosylation in health and disease, and ends with a focus on recent approaches to target N-glycans in rheumatoid arthritis and cancer metastasis.
Collapse
Affiliation(s)
- Sally Esmail
- Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada.
| | - Morris F Manolson
- Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada
| |
Collapse
|
7
|
Cao W, Guo Y, Cheng Z, Xu G, Zuo Q, Nie L, Huang Y, Liu S, Zhu Y. Inducible ATP1B1 Upregulates Antiviral Innate Immune Responses by the Ubiquitination of TRAF3 and TRAF6. THE JOURNAL OF IMMUNOLOGY 2021; 206:2668-2681. [PMID: 34011520 DOI: 10.4049/jimmunol.2001262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/19/2021] [Indexed: 11/19/2022]
Abstract
The antiviral innate immune responses are crucial steps during host defense and must be strictly regulated, but the molecular mechanisms of control remain unclear. In this study, we report increased expression of human ATPase Na+/K+ transporting subunit β 1(ATP1B1) after DNA and RNA virus infections. We found that the expression of ATP1B1 can inhibit viral replication and increase the levels of IFNs, IFN-stimulated genes, and inflammatory cytokines. Knockdown of ATP1B1 by specific short hairpin RNA had the opposite effects. Upon viral infection, ATP1B1 was induced, interacted with TRAF3 and TRAF6, and potentiated the ubiquitination of these proteins, leading to increased phosphorylation of downstream molecules, including TGF-β-activated kinase 1 (TAK1) and TANK-binding kinase 1 (TBK1). These results reveal a previously unrecognized role of ATP1B1 in antiviral innate immunity and suggest a novel mechanism for the induction of IFNs and proinflammatory cytokines during viral infection.
Collapse
Affiliation(s)
- Wei Cao
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yifei Guo
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhikui Cheng
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Gang Xu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Qi Zuo
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Longyu Nie
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yu Huang
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Shi Liu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Ying Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
8
|
Silva CID, Gonçalves-de-Albuquerque CF, Moraes BPTD, Garcia DG, Burth P. Na/K-ATPase: Their role in cell adhesion and migration in cancer. Biochimie 2021; 185:1-8. [PMID: 33713729 DOI: 10.1016/j.biochi.2021.03.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/18/2022]
Abstract
Na/K-ATPase (NKA) is a p-type transmembrane enzyme formed by three different subunits (α, β, and γ gamma). Primarily responsible for transporting sodium and potassium through the cell membrane, it also plays a critical role in intracellular signaling. The activation of diverse intracellular pathways may trigger cell death, survival, or even cell proliferation. Changes in the NKA functions or expression in isoforms subunits impact pathological conditions, such as cancer. The NKA function affects cell adhesion, motility, and migration, which are different in the physiological and pathological states. All enzyme subunits take part in the cell adhesion process, with the β subunit being the most studied. Thus, herein we aim to highlight NKA' central role in cell adhesion, motility, and migration in cancer cells.
Collapse
Affiliation(s)
- Camila Ignácio da Silva
- Laboratório de Enzimologia e Sinalização Celular, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil; Pós-Graduação em Ciências e Biotecnologia Universidade Federal Fluminense, Niterói, Brazil
| | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil; Laboratorio de Imunofarmacologia, Departamento de Bioquímica, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Bianca Portugal Tavares de Moraes
- Laboratorio de Imunofarmacologia, Departamento de Bioquímica, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Diogo Gomes Garcia
- Laboratório de Neurociências Translacional, Programa de Pós-Graduação em Neurologia, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Patrícia Burth
- Laboratório de Enzimologia e Sinalização Celular, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil; Pós-Graduação em Ciências e Biotecnologia Universidade Federal Fluminense, Niterói, Brazil.
| |
Collapse
|
9
|
Medina-Ortiz K, López-Alvarez D, Navia F, Hansen T, Fierro L, Castaño S. Identification of Na +/K +-ATPase α/β isoforms in Rhinella marina tissues by RNAseq and a molecular docking approach at the protein level to evaluate α isoform affinities for bufadienolides. Comp Biochem Physiol A Mol Integr Physiol 2021; 254:110906. [PMID: 33476762 DOI: 10.1016/j.cbpa.2021.110906] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/12/2021] [Accepted: 01/12/2021] [Indexed: 12/24/2022]
Abstract
Na+/K+-ATPase (NKA) function is inhibited by Bufadienolides (BD), a group of cardiotonic steroids (CTS) primarily produced by anurans of the Bufonidae family, such as Rhinella marina. This study characterized the presence of α and β NKA subunit isoforms in R. marina via RNAseq in four tissues: oocytes, skin, heart, and skeletal muscle. Transcripts encoding three α-like isoforms (α1, α2, α3) and three β-like isoforms (β1, β2, β4) were identified. The amino acid sequence of α1-like isoform shared 99.4% identity with the α1 isoform previously published for R. marina. Sequences for α2, α3, and β4 from R. marina were previously unavailable. The first extracellular loop in the α2-like isoform in R. marina showed similar substitutions to those found in their susceptible homologues in other taxa (L/Q111T and S119T); in contrast, this same loop in α3-like isoform showed similar substitutions (Q111L and G120R) to those reported for toad-eating animals such as snakes, which suggests relatively lower affinity for CTS. Docking results showed that all three α-like isoforms identified in R. marina transcriptomes have low affinity to CTS compared to the susceptible α1 isoform of Sus scrofa (pig), with α1-like isoform being the most resistant. The tissue-specific RNAseq results showed the following expression of NKA α-like and β-like subunit isoforms: Oocytes expressed α1 and β1; skin α1, β1, and low levels of β2; heart α1, α3, and β1; skeletal muscle α1, β4, with low levels of α2, α3, and β1. R. marina could be used as an important model for future structural, functional and pharmacological studies of NKA and its isoforms.
Collapse
Affiliation(s)
- Katherine Medina-Ortiz
- Laboratorio de Herpetología y Toxinología, Department of Physiological Sciences, Universidad del Valle, Cali, Colombia.
| | - Diana López-Alvarez
- Laboratorio de Herpetología y Toxinología, Department of Physiological Sciences, Universidad del Valle, Cali, Colombia
| | - Felipe Navia
- Laboratorio de Herpetología y Toxinología, Department of Physiological Sciences, Universidad del Valle, Cali, Colombia
| | - Thomas Hansen
- Laboratorio de Herpetología y Toxinología, Department of Physiological Sciences, Universidad del Valle, Cali, Colombia
| | - Leonardo Fierro
- Laboratorio de Herpetología y Toxinología, Department of Physiological Sciences, Universidad del Valle, Cali, Colombia
| | - Santiago Castaño
- Laboratorio de Herpetología y Toxinología, Department of Physiological Sciences, Universidad del Valle, Cali, Colombia.
| |
Collapse
|
10
|
Marcus EA, Tokhtaeva E, Jimenez JL, Wen Y, Naini BV, Heard AN, Kim S, Capri J, Cohn W, Whitelegge JP, Vagin O. Helicobacter pylori infection impairs chaperone-assisted maturation of Na-K-ATPase in gastric epithelium. Am J Physiol Gastrointest Liver Physiol 2020; 318:G931-G945. [PMID: 32174134 PMCID: PMC7272721 DOI: 10.1152/ajpgi.00266.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 01/31/2023]
Abstract
Helicobacter pylori infection always induces gastritis, which may progress to ulcer disease or cancer. The mechanisms underlying mucosal injury by the bacteria are incompletely understood. Here, we identify a novel pathway for H. pylori-induced gastric injury, the impairment of maturation of the essential transport enzyme and cell adhesion molecule, Na-K-ATPase. Na-K-ATPase comprises α- and β-subunits that assemble in the endoplasmic reticulum (ER) before trafficking to the plasma membrane. Attachment of H. pylori to gastric epithelial cells increased Na-K-ATPase ubiquitylation, decreased its surface and total levels, and impaired ion balance. H. pylori did not alter degradation of plasmalemma-resident Na-K-ATPase subunits or their mRNA levels. Infection decreased association of α- and β-subunits with ER chaperone BiP and impaired assembly of α/β-heterodimers, as was revealed by quantitative mass spectrometry and immunoblotting of immunoprecipitated complexes. The total level of BiP was not altered, and the decrease in interaction with BiP was not observed for other BiP client proteins. The H. pylori-induced decrease in Na-K-ATPase was prevented by BiP overexpression, stopping protein synthesis, or inhibiting proteasomal, but not lysosomal, protein degradation. The results indicate that H. pylori impairs chaperone-assisted maturation of newly made Na-K-ATPase subunits in the ER independently of a generalized ER stress and induces their ubiquitylation and proteasomal degradation. The decrease in Na-K-ATPase levels is also seen in vivo in the stomachs of gerbils and chronically infected children. Further understanding of H. pylori-induced Na-K-ATPase degradation will provide insights for protection against advanced disease.NEW & NOTEWORTHY This work provides evidence that Helicobacter pylori decreases levels of Na-K-ATPase, a vital transport enzyme, in gastric epithelia, both in acutely infected cultured cells and in chronically infected patients and animals. The bacteria interfere with BiP-assisted folding of newly-made Na-K-ATPase subunits in the endoplasmic reticulum, accelerating their ubiquitylation and proteasomal degradation and decreasing efficiency of the assembly of native enzyme. Decreased Na-K-ATPase expression contributes to H. pylori-induced gastric injury.
Collapse
Affiliation(s)
- Elizabeth A Marcus
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, California
- Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California
| | - Elmira Tokhtaeva
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, California
- Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California
| | - Jossue L Jimenez
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, California
- Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California
| | - Yi Wen
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, California
- Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California
| | - Bita V Naini
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Ashley N Heard
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, California
- Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California
| | - Samuel Kim
- Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California
| | - Joseph Capri
- Pasarow Mass Spectrometry Laboratory, The Neuropsychiatric Insititute-Semel Institute, University of California, Los Angeles, California
| | - Whitaker Cohn
- Pasarow Mass Spectrometry Laboratory, The Neuropsychiatric Insititute-Semel Institute, University of California, Los Angeles, California
| | - Julian P Whitelegge
- Pasarow Mass Spectrometry Laboratory, The Neuropsychiatric Insititute-Semel Institute, University of California, Los Angeles, California
| | - Olga Vagin
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, California
- Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California
| |
Collapse
|
11
|
Couto N, Al-Majdoub ZM, Gibson S, Davies PJ, Achour B, Harwood MD, Carlson G, Barber J, Rostami-Hodjegan A, Warhurst G. Quantitative Proteomics of Clinically Relevant Drug-Metabolizing Enzymes and Drug Transporters and Their Intercorrelations in the Human Small Intestine. Drug Metab Dispos 2020; 48:245-254. [PMID: 31959703 PMCID: PMC7076527 DOI: 10.1124/dmd.119.089656] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/23/2019] [Indexed: 01/02/2023] Open
Abstract
The levels of drug-metabolizing enzymes (DMEs) and transporter proteins in the human intestine are pertinent to determine oral drug bioavailability. Despite the paucity of reports on such measurements, it is well recognized that these values are essential for translating in vitro data on drug metabolism and transport to predict drug disposition in gut wall. In the current study, clinically relevant DMEs [cytochrome P450 (P450) and uridine 5'-diphospho-glucuronosyltransferase (UGT)] and drug transporters were quantified in total mucosal protein preparations from the human jejunum (n = 4) and ileum (n = 12) using quantification concatemer-based targeted proteomics. In contrast to previous reports, UGT2B15 and organic anion-transporting polypeptide 1 (OATP1A2) were quantifiable in all our samples. Overall, no significant disparities in protein expression were observed between jejunum and ileum. Relative mRNA expression for drug transporters did not correlate with the abundance of their cognate protein, except for P-glycoprotein 1 (P-gp) and organic solute transporter subunit alpha (OST-α), highlighting the limitations of RNA as a surrogate for protein expression in dynamic tissues with high turnover. Intercorrelations were found within P450 [2C9-2C19 (P = 0.002, R 2 = 0.63), 2C9-2J2 (P = 0.004, R 2 = 0.40), 2D6-2J2 (P = 0.002, R 2 = 0.50)] and UGT [1A1-2B7 (P = 0.02, R 2 = 0.87)] family of enzymes. There were also correlations between P-gp and several other proteins [OST-α (P < 0.0001, R 2 = 0.77), UGT1A6 (P = 0.009, R 2 = 0.38), and CYP3A4 (P = 0.007, R 2 = 0.30)]. Incorporating such correlations into building virtual populations is crucial for obtaining plausible characteristics of simulated individuals. SIGNIFICANCE STATEMENT: A number of drug transporters were quantified for the first time in this study. Several intercorrelations of protein abundance were reported. mRNA expression levels proved to be a poor reflection of differences between individuals regarding the level of protein expression in gut. The reported abundance of drug-metabolizing enzymes and transporters and their intercorrelations will contribute to better predictions of oral drug bioavailability and drug-drug interactions by linking in vitro observations to potential outcomes through physiologically based pharmacokinetic models.
Collapse
Affiliation(s)
- Narciso Couto
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Stephanie Gibson
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Pamela J Davies
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Brahim Achour
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Matthew D Harwood
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Gordon Carlson
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Geoffrey Warhurst
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| |
Collapse
|
12
|
A Model for the Homotypic Interaction between Na +,K +-ATPase β 1 Subunits Reveals the Role of Extracellular Residues 221-229 in Its Ig-Like Domain. Int J Mol Sci 2019; 20:ijms20184538. [PMID: 31540261 PMCID: PMC6770782 DOI: 10.3390/ijms20184538] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/13/2019] [Accepted: 08/16/2019] [Indexed: 12/15/2022] Open
Abstract
The Na+, K+-ATPase transports Na+ and K+ across the membrane of all animal cells. In addition to its ion transporting function, the Na+, K+-ATPase acts as a homotypic epithelial cell adhesion molecule via its β1 subunit. The extracellular region of the Na+, K+-ATPase β1 subunit includes a single globular immunoglobulin-like domain. We performed Molecular Dynamics simulations of the ectodomain of the β1 subunit and a refined protein-protein docking prediction. Our results show that the β1 subunit Ig-like domain maintains an independent structure and dimerizes in an antiparallel fashion. Analysis of the putative interface identified segment Lys221-Tyr229. We generated triple mutations on YFP-β1 subunit fusion proteins to assess the contribution of these residues. CHO fibroblasts transfected with mutant β1 subunits showed a significantly decreased cell-cell adhesion. Association of β1 subunits in vitro was also reduced, as determined by pull-down assays. Altogether, we conclude that two Na+, K+-ATPase molecules recognize each other by a large interface spanning residues 221–229 and 198–207 on their β1 subunits.
Collapse
|
13
|
Litan A, Li Z, Tokhtaeva E, Kelly P, Vagin O, Langhans SA. A Functional Interaction Between Na,K-ATPase β 2-Subunit/AMOG and NF2/Merlin Regulates Growth Factor Signaling in Cerebellar Granule Cells. Mol Neurobiol 2019; 56:7557-7571. [PMID: 31062247 DOI: 10.1007/s12035-019-1592-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 04/02/2019] [Indexed: 10/26/2022]
Abstract
The Na,K-ATPase, consisting of a catalytic α-subunit and a regulatory β-subunit, is a ubiquitously expressed ion pump that carries out the transport of Na+ and K+ across the plasma membranes of most animal cells. In addition to its pump function, Na,K-ATPase serves as a signaling scaffold and a cell adhesion molecule. Of the three β-subunit isoforms, β1 is found in almost all tissues, while β2 expression is mostly restricted to brain and muscle. In cerebellar granule cells, the β2-subunit, also known as adhesion molecule on glia (AMOG), has been linked to neuron-astrocyte adhesion and granule cell migration, suggesting its role in cerebellar development. Nevertheless, little is known about molecular pathways that link the β2-subunit to its cellular functions. Using cerebellar granule precursor cells, we found that the β2-subunit, but not the β1-subunit, negatively regulates the expression of a key activator of the Hippo/YAP signaling pathway, Merlin/neurofibromin-2 (NF2). The knockdown of the β2-subunit resulted in increased Merlin/NF2 expression and affected downstream targets of Hippo signaling, i.e., increased YAP phosphorylation and decreased expression of N-Ras. Further, the β2-subunit knockdown altered the kinetics of epidermal growth factor receptor (EGFR) signaling in a Merlin-dependent mode and impaired EGF-induced reorganization of the actin cytoskeleton. Therefore, our studies for the first time provide a functional link between the Na,K-ATPase β2-subunit and Merlin/NF2 and suggest a role for the β2-subunit in regulating cytoskeletal dynamics and Hippo/YAP signaling during neuronal differentiation.
Collapse
Affiliation(s)
- Alisa Litan
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, DuPont Experimental Station, Bldg 400, #4414, 200 Powder Mill Road, Wilmington, DE, 19803, USA.,Biological Sciences Graduate Program, University of Delaware, Newark, DE, 19716, USA
| | - Zhiqin Li
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, DuPont Experimental Station, Bldg 400, #4414, 200 Powder Mill Road, Wilmington, DE, 19803, USA
| | - Elmira Tokhtaeva
- David Geffen School of Medicine, University of California, Los Angeles, and VA Greater Los Angeles Health Care System, Los Angeles, CA, 90073, USA
| | - Patience Kelly
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, DuPont Experimental Station, Bldg 400, #4414, 200 Powder Mill Road, Wilmington, DE, 19803, USA.,Biological Sciences Graduate Program, University of Delaware, Newark, DE, 19716, USA
| | - Olga Vagin
- David Geffen School of Medicine, University of California, Los Angeles, and VA Greater Los Angeles Health Care System, Los Angeles, CA, 90073, USA
| | - Sigrid A Langhans
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, DuPont Experimental Station, Bldg 400, #4414, 200 Powder Mill Road, Wilmington, DE, 19803, USA.
| |
Collapse
|
14
|
Ouabain Accelerates Collective Cell Migration Through a cSrc and ERK1/2 Sensitive Metalloproteinase Activity. J Membr Biol 2019; 252:549-559. [PMID: 31041466 DOI: 10.1007/s00232-019-00066-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 04/17/2019] [Indexed: 10/26/2022]
Abstract
Studies made in the Madin-Darby canine kidney (MDCK) epithelial cell line showed that ouabain regulates cell adhesion and cell-adhesion-related biological processes, such as migration. Here, we demonstrated that 10 nM ouabain accelerates collective cell migration and heals wounds in cultured MDCK cell monolayers. Ouabain-induced acceleration of cell migration depends on activation of the cSrc-ERK1/2 signaling cascade, as it was inhibited by the kinase inhibitors PP2 and PD98059. Activation of the cSrc-ERK1/2 signaling cascade increased expression and activation of the extracellular matrix metalloproteinase-2 (MMP-2). Inhibition of MMP activity using the generic inhibitor GM6001 or the potent iMMP-2 inhibitor prevented the accelerative effect of ouabain. Likewise, Focal Adhesion Kinase (FAK) inhibition with the transfection of dominant negative peptide FRNK impaired the effect of ouabain. These results suggest that ouabain binding to the Na+,K+-ATPase accelerates collective migration of MDCK cells through activation of the cSrc-ERK1/2-FAK signaling cascade and promoting secretion and MMP activity.
Collapse
|
15
|
Vilchis-Nestor CA, Roldán ML, Leonardi A, Navea JG, Padilla-Benavides T, Shoshani L. Ouabain Enhances Cell-Cell Adhesion Mediated by β 1 Subunits of the Na +,K +-ATPase in CHO Fibroblasts. Int J Mol Sci 2019; 20:E2111. [PMID: 31035668 PMCID: PMC6539428 DOI: 10.3390/ijms20092111] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 12/19/2022] Open
Abstract
Adhesion is a crucial characteristic of epithelial cells to form barriers to pathogens and toxic substances from the environment. Epithelial cells attach to each other using intercellular junctions on the lateral membrane, including tight and adherent junctions, as well as the Na+,K+-ATPase. Our group has shown that non-adherent chinese hamster ovary (CHO) cells transfected with the canine β1 subunit become adhesive, and those homotypic interactions amongst β1 subunits of the Na+,K+-ATPase occur between neighboring epithelial cells. Ouabain, a cardiotonic steroid, binds to the α subunit of the Na+,K+-ATPase, inhibits the pump activity and induces the detachment of epithelial cells when used at concentrations above 300 nM. At nanomolar non-inhibiting concentrations, ouabain affects the adhesive properties of epithelial cells by inducing the expression of cell adhesion molecules through the activation of signaling pathways associated with the α subunit. In this study, we investigated whether the adhesion between β1 subunits was also affected by ouabain. We used CHO fibroblasts stably expressing the β1 subunit of the Na+,K+-ATPase (CHO β1), and studied the effect of ouabain on cell adhesion. Aggregation assays showed that ouabain increased the adhesion between CHO β1 cells. Immunofluorescence and biotinylation assays showed that ouabain (50 nM) increases the expression of the β1 subunit of the Na+,K+-ATPase at the cell membrane. We also examined the effect of ouabain on the activation of signaling pathways in CHO β1 cells, and their subsequent effect on cell adhesion. We found that cSrc is activated by ouabain and, therefore, that it likely regulates the adhesive properties of CHO β1 cells. Collectively, our findings suggest that the β1 subunit adhesion is modulated by the expression levels of the Na+,K+-ATPase at the plasma membrane, which is regulated by ouabain.
Collapse
Affiliation(s)
- Claudia Andrea Vilchis-Nestor
- Department of Physiology Biophysics and Neurosciences, Center for Research and Advanced Studies, Cinvestav-Ipn, CDMX 07360, Mexico.
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - María Luisa Roldán
- Department of Physiology Biophysics and Neurosciences, Center for Research and Advanced Studies, Cinvestav-Ipn, CDMX 07360, Mexico.
| | - Angelina Leonardi
- Department of Chemistry, Skidmore College, 815 North Broadway, Saratoga Springs, NY 12866, USA.
| | - Juan G Navea
- Department of Chemistry, Skidmore College, 815 North Broadway, Saratoga Springs, NY 12866, USA.
| | - Teresita Padilla-Benavides
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Liora Shoshani
- Department of Physiology Biophysics and Neurosciences, Center for Research and Advanced Studies, Cinvestav-Ipn, CDMX 07360, Mexico.
| |
Collapse
|
16
|
Larsen EH, Sørensen JN. Stationary and Nonstationary Ion and Water Flux Interactions in Kidney Proximal Tubule: Mathematical Analysis of Isosmotic Transport by a Minimalistic Model. Rev Physiol Biochem Pharmacol 2019; 177:101-147. [DOI: 10.1007/112_2019_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
AbstractOur mathematical model of epithelial transport (Larsen et al. Acta Physiol. 195:171–186, 2009) is extended by equations for currents and conductance of apical SGLT2. With independent variables of the physiological parameter space, the model reproduces intracellular solute concentrations, ion and water fluxes, and electrophysiology of proximal convoluted tubule. The following were shown:Water flux is given by active Na+flux into lateral spaces, while osmolarity of absorbed fluid depends on osmotic permeability of apical membranes.Following aquaporin “knock-out,” water uptake is not reduced but redirected to the paracellular pathway.Reported decrease in epithelial water uptake in aquaporin-1 knock-out mouse is caused by downregulation of active Na+absorption.Luminal glucose stimulates Na+uptake by instantaneous depolarization-induced pump activity (“cross-talk”) and delayed stimulation because of slow rise in intracellular [Na+].Rate of fluid absorption and flux of active K+absorption would have to be attuned at epithelial cell level for the [K+] of the absorbate being in the physiological range of interstitial [K+].Following unilateral osmotic perturbation, time course of water fluxes between intraepithelial compartments provides physical explanation for the transepithelial osmotic permeability being orders of magnitude smaller than cell membranes’ osmotic permeability.Fluid absorption is always hyperosmotic to bath.Deviation from isosmotic absorption is increased in presence of glucose contrasting experimental studies showing isosmotic transport being independent of glucose uptake.For achieving isosmotic transport, the cost of Na+recirculation is predicted to be but a few percent of the energy consumption of Na+/K+pumps.
Collapse
|
17
|
Rojas M, Díaz P, León P, Gonzalez AA, González M, Barrientos V, Pestov NB, Alzamora R, Michea L. Mineralocorticoids modulate the expression of the β-3 subunit of the Na +, K +-ATPase in the renal collecting duct. Channels (Austin) 2017. [PMID: 28636485 DOI: 10.1080/19336950.2017.1344800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Renal sodium reabsorption depends on the activity of the Na+,K+-ATPase α/β heterodimer. Four α (α1-4) and 3 β (β1-3) subunit isoforms have been described. It is accepted that renal tubule cells express α1/β1 dimers. Aldosterone stimulates Na+,K+-ATPase activity and may modulate α1/β1 expression. However, some studies suggest the presence of β3 in the kidney. We hypothesized that the β3 isoform of the Na+,K+-ATPase is expressed in tubular cells of the distal nephron, and modulated by mineralocorticoids. We found that β3 is highly expressed in collecting duct of rodents, and that mineralocorticoids decreased the expression of β3. Thus, we describe a novel molecular mechanism of sodium pump modulation that may contribute to the effects of mineralocorticoids on sodium reabsorption.
Collapse
Affiliation(s)
- Macarena Rojas
- a Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile
| | - Pablo Díaz
- a Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile
| | - Pablo León
- a Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile
| | - Alexis A Gonzalez
- b Instituto de Química, Pontificia Universidad Católica de Valparaíso , Valparaíso , Chile
| | - Magdalena González
- a Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile
| | - Víctor Barrientos
- a Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile
| | - Nikolay B Pestov
- c Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry , Moscow , Russia.,d Department of Physiology and Pharmacology and Center for Diabetes and Endocrine Research , University of Toledo College of Medicine , Toledo , OH , USA
| | - Rodrigo Alzamora
- a Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile.,e Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD) , Santiago , Chile
| | - Luis Michea
- a Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile.,f Millennium Institute on Immunology and Immunotherapy , Santiago , Chile
| |
Collapse
|
18
|
Mazel T. Crosstalk of cell polarity signaling pathways. PROTOPLASMA 2017; 254:1241-1258. [PMID: 28293820 DOI: 10.1007/s00709-017-1075-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/02/2017] [Indexed: 06/06/2023]
Abstract
Cell polarity, the asymmetric organization of cellular components along one or multiple axes, is present in most cells. From budding yeast cell polarization induced by pheromone signaling, oocyte polarization at fertilization to polarized epithelia and neuronal cells in multicellular organisms, similar mechanisms are used to determine cell polarity. Crucial role in this process is played by signaling lipid molecules, small Rho family GTPases and Par proteins. All these signaling circuits finally govern the cytoskeleton, which is responsible for oriented cell migration, cell shape changes, and polarized membrane and organelle trafficking. Thus, typically in the process of cell polarization, most cellular constituents become polarized, including plasma membrane lipid composition, ion concentrations, membrane receptors, and proteins in general, mRNA, vesicle trafficking, or intracellular organelles. This review gives a brief overview how these systems talk to each other both during initial symmetry breaking and within the signaling feedback loop mechanisms used to preserve the polarized state.
Collapse
Affiliation(s)
- Tomáš Mazel
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Albertov 4, 128 00, Prague 2, Czech Republic.
- State Institute for Drug Control, Šrobárova 48, 100 41, Prague 10, Czech Republic.
| |
Collapse
|
19
|
Caceres PS, Benedicto I, Lehmann GL, Rodriguez-Boulan EJ. Directional Fluid Transport across Organ-Blood Barriers: Physiology and Cell Biology. Cold Spring Harb Perspect Biol 2017; 9:a027847. [PMID: 28003183 PMCID: PMC5334253 DOI: 10.1101/cshperspect.a027847] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Directional fluid flow is an essential process for embryo development as well as for organ and organism homeostasis. Here, we review the diverse structure of various organ-blood barriers, the driving forces, transporters, and polarity mechanisms that regulate fluid transport across them, focusing on kidney-, eye-, and brain-blood barriers. We end by discussing how cross talk between barrier epithelial and endothelial cells, perivascular cells, and basement membrane signaling contribute to generate and maintain organ-blood barriers.
Collapse
Affiliation(s)
- Paulo S Caceres
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| | - Ignacio Benedicto
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| | - Guillermo L Lehmann
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| | - Enrique J Rodriguez-Boulan
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| |
Collapse
|
20
|
Shi JL, Fu L, Ang Q, Wang GJ, Zhu J, Wang WD. Overexpression of ATP1B1 predicts an adverse prognosis in cytogenetically normal acute myeloid leukemia. Oncotarget 2016; 7:2585-95. [PMID: 26506237 PMCID: PMC4823057 DOI: 10.18632/oncotarget.6226] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 10/09/2015] [Indexed: 11/25/2022] Open
Abstract
ATP1B1 encodes the Na,K-ATPase β subunit, a key regulator of the Na+ and K+ electrochemical gradients across the plasma membrane and an essential regulator of cellular activity. We used several microarray datasets to test the prognostic efficacy of ATP1B1 expression in cytogenetically normal acute myeloid leukemia (CN-AML). Within the primary cohort (n = 157), high ATP1B1 expression (ATP1B1high) was associated with shorter overall survival (OS) and event-free survival (EFS) (P = 0.0068, P = 0.0039, respectively). Similar results were also obtained in the European Leukemia Net (ELN) Intermediate-I genetic category (OS: P = 0.0035, EFS: P = 0.0007). Multivariable analyses confirmed ATP1B1high is an independent predictor of shorter OS (P = 0.042) and EFS (P = 0.035). Analysis of another CN-AML cohort confirmed that ATP1B1high is associated with shorter OS (P = 0.0046, n = 162). In addition, up-regulation of oncogenes/onco-microRNAs such as MYCN, CCND2, CDK6, KIT and miR-155, among others, was associated with ATP1B1high, which may be indicative of ATP1B1's leukemogenicity. Our results may improve risk stratification and indicate new therapeutic targets for CN-AML.
Collapse
Affiliation(s)
- Jin-long Shi
- Medical Engineering Support Center, Chinese PLA General Hospital, Beijing, China
| | - Lin Fu
- Department of Hematology and Lymphoma Research Center, Peking University, Third Hospital, Beijing, China
| | - Qing Ang
- Medical Engineering Support Center, Chinese PLA General Hospital, Beijing, China
| | - Guo-jing Wang
- Medical Engineering Support Center, Chinese PLA General Hospital, Beijing, China
| | - Jun Zhu
- Medical Engineering Support Center, Chinese PLA General Hospital, Beijing, China
| | - Wei-dong Wang
- Medical Engineering Support Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
21
|
Lobato-Álvarez JA, Roldán ML, López-Murillo TDC, González-Ramírez R, Bonilla-Delgado J, Shoshani L. The Apical Localization of Na +, K +-ATPase in Cultured Human Retinal Pigment Epithelial Cells Depends on Expression of the β 2 Subunit. Front Physiol 2016; 7:450. [PMID: 27774068 PMCID: PMC5054689 DOI: 10.3389/fphys.2016.00450] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 09/20/2016] [Indexed: 11/28/2022] Open
Abstract
Na+, K+-ATPase, or the Na+ pump, is a key component in the maintenance of the epithelial phenotype. In most epithelia, the pump is located in the basolateral domain. Studies from our laboratory have shown that the β1 subunit of Na+, K+-ATPase plays an important role in this mechanism because homotypic β1-β1 interactions between neighboring cells stabilize the pump in the lateral membrane. However, in the retinal pigment epithelium (RPE), the Na+ pump is located in the apical domain. The mechanism of polarization in this epithelium is unclear. We hypothesized that the apical polarization of the pump in RPE cells depends on the expression of its β2 subunit. ARPE-19 cells cultured for up to 8 weeks on inserts did not polarize, and Na+, K+-ATPase was expressed in the basolateral membrane. In the presence of insulin, transferrin and selenic acid (ITS), ARPE-19 cells cultured for 4 weeks acquired an RPE phenotype, and the Na+ pump was visible in the apical domain. Under these conditions, Western blot analysis was employed to detect the β2 isoform and immunofluorescence analysis revealed an apparent apical distribution of the β2 subunit. qPCR results showed a time-dependent increase in the level of β2 isoform mRNA, suggesting regulation at the transcriptional level. Moreover, silencing the expression of the β2 isoform in ARPE-19 cells resulted in a decrease in the apical localization of the pump, as assessed by the mislocalization of the α2 subunit in that domain. Our results demonstrate that the apical polarization of Na+, K+-ATPase in RPE cells depends on the expression of the β2 subunit.
Collapse
Affiliation(s)
- Jorge A Lobato-Álvarez
- Laboratory of Epithelial Research, Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN México City, Mexico
| | - María L Roldán
- Laboratory of Epithelial Research, Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN México City, Mexico
| | - Teresa Del Carmen López-Murillo
- Laboratory of Epithelial Research, Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN México City, Mexico
| | - Ricardo González-Ramírez
- Department of Molecular Biology and Histocompatibility, Hospital General Dr. Manuel Gea González México City, Mexico
| | - José Bonilla-Delgado
- Research Unit, Laboratory of Genetics and Molecular Diagnosis, Hospital Juárez de México México City, Mexico
| | - Liora Shoshani
- Laboratory of Epithelial Research, Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN México City, Mexico
| |
Collapse
|
22
|
β1-Na(+),K(+)-ATPase gene therapy upregulates tight junctions to rescue lipopolysaccharide-induced acute lung injury. Gene Ther 2016; 23:489-99. [PMID: 26910760 DOI: 10.1038/gt.2016.19] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 01/08/2016] [Accepted: 01/19/2016] [Indexed: 01/05/2023]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are associated with diverse disorders and characterized by disruption of the alveolar-capillary barrier, leakage of edema fluid into the lung, and substantial inflammation leading to acute respiratory failure. Gene therapy is a potentially powerful approach to treat ALI/ARDS through repair of alveolar epithelial function. Herein, we show that delivery of a plasmid expressing β1-subunit of the Na(+),K(+)-ATPase (β1-Na(+),K(+)-ATPase) alone or in combination with epithelial sodium channel (ENaC) α1-subunit using electroporation not only protected from subsequent lipopolysaccharide (LPS)-mediated lung injury, but also treated injured lungs. However, transfer of α1-subunit of ENaC (α1-ENaC) alone only provided protection benefit rather than treatment benefit although alveolar fluid clearance had been remarkably enhanced. Gene transfer of β1-Na(+),K(+)-ATPase, but not α1-ENaC, not only enhanced expression of tight junction protein zona occludins-1 (ZO-1) and occludin both in cultured cells and in mouse lungs, but also reduced pre-existing increase of lung permeability in vivo. These results demonstrate that gene transfer of β1-Na(+),K(+)-ATPase upregulates tight junction formation and therefore treats lungs with existing injury, whereas delivery of α1-ENaC only maintains pre-existing tight junction but not for generation. This indicates that the restoration of epithelial/endothelial barrier function may provide better treatment of ALI/ARDS.
Collapse
|
23
|
Kumar V, Prasad B, Patilea G, Gupta A, Salphati L, Evers R, Hop CECA, Unadkat JD. Quantitative transporter proteomics by liquid chromatography with tandem mass spectrometry: addressing methodologic issues of plasma membrane isolation and expression-activity relationship. Drug Metab Dispos 2015; 43:284-8. [PMID: 25488931 DOI: 10.1124/dmd.114.061614] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
To predict transporter-mediated drug disposition using physiologically based pharmacokinetic models, one approach is to measure transport activity and relate it to protein expression levels in cell lines (overexpressing the transporter) and then scale these to via in vitro to in vivo extrapolation (IVIVE). This approach makes two major assumptions. First, that the expression of the transporter is predominantly in the plasma membrane. Second, that there is a linear correlation between expression level and activity of the transporter protein. The present study was conducted to test these two assumptions. We evaluated two commercially available kits that claimed to separate plasma membrane from other cell membranes. The Qiagen Qproteome kit yielded very little protein in the fraction purported to be the plasma membrane. The Abcam Phase Separation kit enriched the plasma membrane but did not separate it from other intracellular membranes. For the Abcam method, the expression level of organic anion-transporting polypeptides (OATP) 1B1/2B1 and breast cancer resistance protein (BCRP) proteins in all subcellular fractions isolated from cells or human liver tissue tracked that of Na⁺-K⁺ ATPase. Assuming that Na⁺-K⁺ ATPase is predominantly located in the plasma membrane, these data suggest that the transporters measured are also primarily located in the plasma membrane. Using short hairpin RNA, we created clones of cell lines with varying degrees of OATP1B1 or BCRP expression level. In these clones, transport activity of OATP1B1 or BCRP was highly correlated with protein expression level (r² > 0.9). These data support the use of transporter expression level data and activity data from transporter overexpressing cell lines for IVIVE of transporter-mediated disposition of drugs.
Collapse
Affiliation(s)
- Vineet Kumar
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., B.P., G.P., J.D.U.); Drug Metabolism and Pharmacokinetics, Infection Innovative Medicines Unit, AstraZeneca Pharmaceuticals LLP, Waltham, Massachusetts (A.G.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (R.E.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, Californai (L.S., C.E.C.A.H.)
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., B.P., G.P., J.D.U.); Drug Metabolism and Pharmacokinetics, Infection Innovative Medicines Unit, AstraZeneca Pharmaceuticals LLP, Waltham, Massachusetts (A.G.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (R.E.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, Californai (L.S., C.E.C.A.H.)
| | - Gabriela Patilea
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., B.P., G.P., J.D.U.); Drug Metabolism and Pharmacokinetics, Infection Innovative Medicines Unit, AstraZeneca Pharmaceuticals LLP, Waltham, Massachusetts (A.G.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (R.E.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, Californai (L.S., C.E.C.A.H.)
| | - Anshul Gupta
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., B.P., G.P., J.D.U.); Drug Metabolism and Pharmacokinetics, Infection Innovative Medicines Unit, AstraZeneca Pharmaceuticals LLP, Waltham, Massachusetts (A.G.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (R.E.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, Californai (L.S., C.E.C.A.H.)
| | - Laurent Salphati
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., B.P., G.P., J.D.U.); Drug Metabolism and Pharmacokinetics, Infection Innovative Medicines Unit, AstraZeneca Pharmaceuticals LLP, Waltham, Massachusetts (A.G.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (R.E.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, Californai (L.S., C.E.C.A.H.)
| | - Raymond Evers
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., B.P., G.P., J.D.U.); Drug Metabolism and Pharmacokinetics, Infection Innovative Medicines Unit, AstraZeneca Pharmaceuticals LLP, Waltham, Massachusetts (A.G.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (R.E.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, Californai (L.S., C.E.C.A.H.)
| | - Cornelis E C A Hop
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., B.P., G.P., J.D.U.); Drug Metabolism and Pharmacokinetics, Infection Innovative Medicines Unit, AstraZeneca Pharmaceuticals LLP, Waltham, Massachusetts (A.G.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (R.E.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, Californai (L.S., C.E.C.A.H.)
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., B.P., G.P., J.D.U.); Drug Metabolism and Pharmacokinetics, Infection Innovative Medicines Unit, AstraZeneca Pharmaceuticals LLP, Waltham, Massachusetts (A.G.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (R.E.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, Californai (L.S., C.E.C.A.H.)
| |
Collapse
|
24
|
Tokhtaeva E, Capri J, Marcus EA, Whitelegge JP, Khuzakhmetova V, Bukharaeva E, Deiss-Yehiely N, Dada LA, Sachs G, Fernandez-Salas E, Vagin O. Septin dynamics are essential for exocytosis. J Biol Chem 2015; 290:5280-97. [PMID: 25575596 DOI: 10.1074/jbc.m114.616201] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Septins are a family of 14 cytoskeletal proteins that dynamically form hetero-oligomers and organize membrane microdomains for protein complexes. The previously reported interactions with SNARE proteins suggested the involvement of septins in exocytosis. However, the contradictory results of up- or down-regulation of septin-5 in various cells and mouse models or septin-4 in mice suggested either an inhibitory or a stimulatory role for these septins in exocytosis. The involvement of the ubiquitously expressed septin-2 or general septin polymerization in exocytosis has not been explored to date. Here, by nano-LC with tandem MS and immunoblot analyses of the septin-2 interactome in mouse brain, we identified not only SNARE proteins but also Munc-18-1 (stabilizes assembled SNARE complexes), N-ethylmaleimide-sensitive factor (NSF) (disassembles SNARE complexes after each membrane fusion event), and the chaperones Hsc70 and synucleins (maintain functional conformation of SNARE proteins after complex disassembly). Importantly, α-soluble NSF attachment protein (SNAP), the adaptor protein that mediates NSF binding to the SNARE complex, did not interact with septin-2, indicating that septins undergo reorganization during each exocytosis cycle. Partial depletion of septin-2 by siRNA or impairment of septin dynamics by forchlorfenuron inhibited constitutive and stimulated exocytosis of secreted and transmembrane proteins in various cell types. Forchlorfenuron impaired the interaction between SNAP-25 and its chaperone Hsc70, decreasing SNAP-25 levels in cultured neuroendocrine cells, and inhibited both spontaneous and stimulated acetylcholine secretion in mouse motor neurons. The results demonstrate a stimulatory role of septin-2 and the dynamic reorganization of septin oligomers in exocytosis.
Collapse
Affiliation(s)
- Elmira Tokhtaeva
- From the Departments of Physiology and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California 90073
| | - Joe Capri
- The Neuropsychiatric Institute-Semel Institute, Pasarow Mass Spectrometry Laboratory, UCLA, Los Angeles, California 90024
| | - Elizabeth A Marcus
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California 90073, Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California 90095
| | - Julian P Whitelegge
- The Neuropsychiatric Institute-Semel Institute, Pasarow Mass Spectrometry Laboratory, UCLA, Los Angeles, California 90024
| | - Venera Khuzakhmetova
- Kazan Institute of Biochemistry and Biophysics, Kazan Scientific Center of the Russian Academy of Sciences, Kazan 420111, Russia, Kazan Federal University, Kazan 420008, Russia
| | - Ellya Bukharaeva
- Kazan Institute of Biochemistry and Biophysics, Kazan Scientific Center of the Russian Academy of Sciences, Kazan 420111, Russia, Kazan Federal University, Kazan 420008, Russia
| | - Nimrod Deiss-Yehiely
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, and
| | - Laura A Dada
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, and
| | - George Sachs
- From the Departments of Physiology and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California 90073
| | - Ester Fernandez-Salas
- Department of Pathology, School of Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | - Olga Vagin
- From the Departments of Physiology and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California 90073,
| |
Collapse
|
25
|
Rocha SC, Pessoa MTC, Neves LDR, Alves SLG, Silva LM, Santos HL, Oliveira SMF, Taranto AG, Comar M, Gomes IV, Santos FV, Paixão N, Quintas LEM, Noël F, Pereira AF, Tessis ACSC, Gomes NLS, Moreira OC, Rincon-Heredia R, Varotti FP, Blanco G, Villar JAFP, Contreras RG, Barbosa LA. 21-Benzylidene digoxin: a proapoptotic cardenolide of cancer cells that up-regulates Na,K-ATPase and epithelial tight junctions. PLoS One 2014; 9:e108776. [PMID: 25290152 PMCID: PMC4188576 DOI: 10.1371/journal.pone.0108776] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 08/25/2014] [Indexed: 02/07/2023] Open
Abstract
Cardiotonic steroids are used to treat heart failure and arrhythmia and have promising anticancer effects. The prototypic cardiotonic steroid ouabain may also be a hormone that modulates epithelial cell adhesion. Cardiotonic steroids consist of a steroid nucleus and a lactone ring, and their biological effects depend on the binding to their receptor, Na,K-ATPase, through which, they inhibit Na+ and K+ ion transport and activate of several intracellular signaling pathways. In this study, we added a styrene group to the lactone ring of the cardiotonic steroid digoxin, to obtain 21-benzylidene digoxin (21-BD), and investigated the effects of this synthetic cardiotonic steroid in different cell models. Molecular modeling indicates that 21-BD binds to its target Na,K-ATPase with low affinity, adopting a different pharmacophoric conformation when bound to its receptor than digoxin. Accordingly, 21-DB, at relatively high µM amounts inhibits the activity of Na,K-ATPase α1, but not α2 and α3 isoforms. In addition, 21-BD targets other proteins outside the Na,K-ATPase, inhibiting the multidrug exporter Pdr5p. When used on whole cells at low µM concentrations, 21-BD produces several effects, including: 1) up-regulation of Na,K-ATPase expression and activity in HeLa and RKO cancer cells, which is not found for digoxin, 2) cell specific changes in cell viability, reducing it in HeLa and RKO cancer cells, but increasing it in normal epithelial MDCK cells, which is different from the response to digoxin, and 3) changes in cell-cell interaction, altering the molecular composition of tight junctions and elevating transepithelial electrical resistance of MDCK monolayers, an effect previously found for ouabain. These results indicate that modification of the lactone ring of digoxin provides new properties to the compound, and shows that the structural change introduced could be used for the design of cardiotonic steroid with novel functions.
Collapse
Affiliation(s)
- Sayonarah C. Rocha
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Divinópolis, MG, Brazil
| | - Marco T. C. Pessoa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Divinópolis, MG, Brazil
| | - Luiza D. R. Neves
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Divinópolis, MG, Brazil
| | - Silmara L. G. Alves
- Laboratório de Síntese Orgânica, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Divinópolis, MG, Brazil
| | - Luciana M. Silva
- Laboratório de Biologia Celular e Inovação Biotecnológica, Fundação Ezequiel Dias, Belo Horizonte, MG, Brazil
| | - Herica L. Santos
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Divinópolis, MG, Brazil
| | - Soraya M. F. Oliveira
- Laboratório de Bioinformática, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Divinópolis, MG, Brazil
| | - Alex G. Taranto
- Laboratório de Bioinformática, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Divinópolis, MG, Brazil
| | - Moacyr Comar
- Laboratório de Bioinformática, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Divinópolis, MG, Brazil
| | - Isabella V. Gomes
- Laboratório de Biologia Celular e Mutagenicidade, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Divinópolis, MG, Brazil
| | - Fabio V. Santos
- Laboratório de Biologia Celular e Mutagenicidade, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Divinópolis, MG, Brazil
| | - Natasha Paixão
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luis E. M. Quintas
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - François Noël
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Antonio F. Pereira
- Laboratório de Bioquímica Microbiana, Instituto de Microbiologia Paulo Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ana C. S. C. Tessis
- Laboratório de Bioquímica Microbiana, Instituto de Microbiologia Paulo Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro (IFRJ), Rio de Janeiro, RJ, Brazil
| | - Natalia L. S. Gomes
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Otacilio C. Moreira
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Ruth Rincon-Heredia
- Department of Physiology, Biophysics and Neurosciences, Center for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Fernando P. Varotti
- Laboratório de Bioquímica de Parasitos, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Divinópolis, MG, Brazil
| | - Gustavo Blanco
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Jose A. F. P. Villar
- Laboratório de Síntese Orgânica, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Divinópolis, MG, Brazil
| | - Rubén G. Contreras
- Department of Physiology, Biophysics and Neurosciences, Center for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Leandro A. Barbosa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Divinópolis, MG, Brazil
| |
Collapse
|
26
|
Ouabain induces endocytosis and degradation of tight junction proteins through ERK1/2-dependent pathways. Exp Cell Res 2014; 320:108-18. [DOI: 10.1016/j.yexcr.2013.10.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 10/07/2013] [Accepted: 10/09/2013] [Indexed: 11/20/2022]
|
27
|
|
28
|
Vagin O, Dada LA, Tokhtaeva E, Sachs G. The Na-K-ATPase α₁β₁ heterodimer as a cell adhesion molecule in epithelia. Am J Physiol Cell Physiol 2012; 302:C1271-81. [PMID: 22277755 PMCID: PMC3361946 DOI: 10.1152/ajpcell.00456.2011] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Accepted: 01/24/2012] [Indexed: 11/22/2022]
Abstract
The ion gradients generated by the Na-K-ATPase play a critical role in epithelia by driving transepithelial transport of various solutes. The efficiency of this Na-K-ATPase-driven vectorial transport depends on the integrity of epithelial junctions that maintain polar distribution of membrane transporters, including the basolateral sodium pump, and restrict paracellular diffusion of solutes. The review summarizes the data showing that, in addition to pumping ions, the Na-K-ATPase located at the sites of cell-cell junction acts as a cell adhesion molecule by interacting with the Na-K-ATPase of the adjacent cell in the intercellular space accompanied by anchoring to the cytoskeleton in the cytoplasm. The review also discusses the experimental evidence on the importance of a specific amino acid region in the extracellular domain of the Na-K-ATPase β(1) subunit for the Na-K-ATPase trans-dimerization and intercellular adhesion. Furthermore, a possible role of N-glycans linked to the Na-K-ATPase β(1) subunit in regulation of epithelial junctions by modulating β(1)-β(1) interactions is discussed.
Collapse
Affiliation(s)
- Olga Vagin
- Department of Physiology, School of Medicine, University of California Los Angeles and Veterans Administration Greater Los Angeles Health Care System, Los Angeles, California, USA.
| | | | | | | |
Collapse
|
29
|
Tokhtaeva E, Sachs G, Sun H, Dada LA, Sznajder JI, Vagin O. Identification of the amino acid region involved in the intercellular interaction between the β1 subunits of Na+/K+ -ATPase. J Cell Sci 2012; 125:1605-16. [PMID: 22328500 DOI: 10.1242/jcs.100149] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Epithelial junctions depend on intercellular interactions between β(1) subunits of the Na(+)/K(+)-ATPase molecules of neighboring cells. The interaction between dog and rat subunits is less effective than the interaction between two dog β(1) subunits, indicating the importance of species-specific regions for β(1)-β(1) binding. To identify these regions, the species-specific amino acid residues were mapped on a high-resolution structure of the Na(+)/K(+)-ATPase β(1) subunit to select those exposed towards the β(1) subunit of the neighboring cell. These exposed residues were mutated in both dog and rat YFP-linked β(1) subunits (YFP-β(1)) and also in the secreted extracellular domain of the dog β(1) subunit. Five rat-like mutations in the amino acid region spanning residues 198-207 of the dog YFP-β(1) expressed in Madin-Darby canine kidney (MDCK) cells decreased co-precipitation of the endogenous dog β(1) subunit with YFP-β(1) to the level observed between dog β(1) and rat YFP-β(1). In parallel, these mutations impaired the recognition of YFP-β(1) by the dog-specific antibody that inhibits cell adhesion between MDCK cells. Accordingly, dog-like mutations in rat YFP-β(1) increased both the (YFP-β(1))-β(1) interaction in MDCK cells and recognition by the antibody. Conversely, rat-like mutations in the secreted extracellular domain of the dog β(1) subunit increased its interaction with rat YFP-β(1) in vitro. In addition, these mutations resulted in a reduction of intercellular adhesion between rat lung epithelial cells following addition of the secreted extracellular domain of the dog β(1) subunit to a cell suspension. Therefore, the amino acid region 198-207 is crucial for both trans-dimerization of the Na(+)/K(+)-ATPase β(1) subunits and cell-cell adhesion.
Collapse
Affiliation(s)
- Elmira Tokhtaeva
- Department of Physiology, School of Medicine, UCLA and Veterans Administration Greater Los Angeles Health Care System, Los Angeles, VAGLAHS/West LA, Building 113, Room 324, 11301 Wilshire Boulevard, Los Angeles, CA 90073, USA
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
The exchange of substances between higher organisms and the environment occurs across transporting epithelia whose basic features are tight junctions (TJs) that seal the intercellular space, and polarity, which enables cells to transport substances vectorially. In a previous study, we demonstrated that 10 nM ouabain modulates TJs, and we now show that it controls polarity as well. We gauge polarity through the development of a cilium at the apical domain of Madin-Darby canine kidney cells (MDCK, epithelial dog kidney). Ouabain accelerates ciliogenesis in an ERK1/2-dependent manner. Claudin-2, a molecule responsible for the Na(+) and H(2)O permeability of the TJs, is also present at the cilium, as it colocalizes and coprecipitates with acetylated α-tubulin. Ouabain modulates claudin-2 localization at the cilium through ERK1/2. Comparing wild-type and ouabain-resistant MDCK cells, we show that ouabain acts through Na(+),K(+)-ATPase. Taken together, our previous and present results support the possibility that ouabain constitutes a hormone that modulates the transporting epithelial phenotype, thereby playing a crucial role in metazoan life.
Collapse
|
31
|
Cereijido M, Contreras RG, Shoshani L, Larre I. The Na+-K+-ATPase as self-adhesion molecule and hormone receptor. Am J Physiol Cell Physiol 2011; 302:C473-81. [PMID: 22049208 DOI: 10.1152/ajpcell.00083.2011] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Thanks to the homeostasis of the internal milieu, metazoan cells can enormously simplify their housekeeping efforts and engage instead in differentiation and multiple forms of organization (tissues, organs, systems) that enable them to produce an astonishing diversity of mammals. The stability of the internal milieu despite drastic variations of the external environment (air, fresh or seawater, gastrointestinal fluids, glomerular filtrate, bile) is due to transporting epithelia that can adjust their specific permeability to H(2)O, H(+), Na(+), K(+), Ca(2+), and Cl(-) over several orders of magnitude and exchange substances with the outer milieu with exquisite precision. This exchange is due to the polarized expression of membrane proteins, among them Na(+)-K(+)-ATPase, an oligomeric enzyme that uses chemical energy from ATP molecules to translocate ions across the plasma membrane of epithelial cells. Na(+)-K(+)-ATPase presents two types of asymmetries: the arrangement of its subunits, and its expression in one pole of the epithelial cell ("polarity"). In most epithelia, polarity consists of the expression of Na(+)-K(+)-ATPase towards the intercellular space and arises in part from the interaction of the extracellular segment of the β-subunit with another β-subunit present in a Na(+)-K(+)-ATPase molecule expressed by a neighboring cell. In addition to enabling the Na(+)-K(+)-ATPase to transport ions and water vectorially, this position exposes its receptors to ouabain and analogous cardiotonic steroids, which are present in the internal milieu because these were secreted by endocrine cells.
Collapse
Affiliation(s)
- M Cereijido
- CINVESTAV, Col. San Pedro Zacatenco, Del. Gustavo A. Madero, México, D.F., México.
| | | | | | | |
Collapse
|
32
|
Liu L, Ivanov AV, Gable ME, Jolivel F, Morrill GA, Askari A. Comparative properties of caveolar and noncaveolar preparations of kidney Na+/K+-ATPase. Biochemistry 2011; 50:8664-73. [PMID: 21905705 PMCID: PMC3186040 DOI: 10.1021/bi2009008] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
![]()
To evaluate previously proposed functions of renal caveolar
Na+/K+-ATPase, we modified the standard procedures
for the preparation of the purified membrane-bound kidney enzyme,
separated the caveolar and noncaveolar pools, and compared their properties.
While the subunits of Na+/K+-ATPase (α,β,γ)
constituted most of the protein content of the noncaveolar pool, the
caveolar pool also contained caveolins and major caveolar proteins
annexin-2 tetramer and E-cadherin. Ouabain-sensitive Na+/K+-ATPase activities of the two pools had similar properties
and equal molar activities, indicating that the caveolar enzyme retains
its ion transport function and does not contain nonpumping enzyme.
As minor constituents, both caveolar and noncaveolar pools also contained
Src, EGFR, PI3K, and several other proteins known to be involved in
stimulous-induced signaling by Na+/K+-ATPase,
indicating that signaling function is not limited to the caveolar
pool. Endogenous Src was active in both pools but was not further
activated by ouabain, calling into question direct interaction of
Src with native Na+/K+-ATPase. Chemical cross-linking,
co-immunoprecipitation, and immunodetection studies showed that in
the caveolar pool, caveolin-1 oligomers, annexin-2 tetramers, and
oligomers of the α,β,γ-protomers of Na+/K+-ATPase form a large multiprotein complex. In conjunction
with known roles of E-cadherin and the β-subunit of Na+/K+-ATPase in cell adhesion and noted intercellular β,β-contacts
within the structure of Na+/K+-ATPase, our findings
suggest that interacting caveolar Na+/K+-ATPases
located at renal adherens junctions maintain contact of two adjacent
cells, conduct essential ion pumping, and are capable of locus-specific
signaling in junctional cells.
Collapse
Affiliation(s)
- Lijun Liu
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, Ohio 43614, United States
| | | | | | | | | | | |
Collapse
|
33
|
Tokhtaeva E, Sachs G, Souda P, Bassilian S, Whitelegge JP, Shoshani L, Vagin O. Epithelial junctions depend on intercellular trans-interactions between the Na,K-ATPase β₁ subunits. J Biol Chem 2011; 286:25801-12. [PMID: 21642423 DOI: 10.1074/jbc.m111.252247] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
N-Glycans of the Na,K-ATPase β₁ subunit are important for intercellular adhesion in epithelia, suggesting that epithelial junctions depend on N-glycan-mediated interactions between the β₁ subunits of neighboring cells. The level of co-immunoprecipitation of the endogenous β₁ subunit with various YFP-linked β₁ subunits expressed in Madin-Darby canine kidney cells was used to assess β₁-β₁ interactions. The amount of co-precipitated endogenous dog β₁ was greater with dog YFP-β₁ than with rat YFP-β₁, showing that amino acid-mediated interactions are important for β₁-β₁ binding. Co-precipitation of β₁ was also less with the unglycosylated YFP-β₁ than with glycosylated YFP-β₁, indicating a role for N-glycans. Mixing cells expressing dog YFP-β₁ with non-transfected cells increased the amount of co-precipitated β₁, confirming the presence of intercellular (YFP-β₁)-β₁ complexes. Accordingly, disruption of intercellular junctions decreased the amount of co-precipitated β₁ subunits. The decrease in β₁ co-precipitation both with rat YFP-β₁ and unglycosylated YFP-β₁ was associated with decreased detergent stability of junctional proteins and increased paracellular permeability. Reducing N-glycan branching by specific inhibitors increased (YFP-β₁)-β₁ co-precipitation and strengthened intercellular junctions. Therefore, interactions between the β₁ subunits of neighboring cells maintain integrity of intercellular junctions, and alterations in the β₁ subunit N-glycan structure can regulate stability and tightness of intercellular junctions.
Collapse
Affiliation(s)
- Elmira Tokhtaeva
- Department of Physiology, School of Medicine, UCLA and Veterans Administration Greater Los Angeles Health Care System, Los Angeles, California 90073, USA
| | | | | | | | | | | | | |
Collapse
|