1
|
Vagin O, Tokhtaeva E, Larauche M, Davood J, Marcus EA. Helicobacter pylori-Induced Decrease in Membrane Expression of Na,K-ATPase Leads to Gastric Injury. Biomolecules 2024; 14:772. [PMID: 39062486 PMCID: PMC11274427 DOI: 10.3390/biom14070772] [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: 05/01/2024] [Revised: 06/12/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Helicobacter pylori is a highly prevalent human gastric pathogen that causes gastritis, ulcer disease, and gastric cancer. It is not yet fully understood how H. pylori injures the gastric epithelium. The Na,K-ATPase, an essential transporter found in virtually all mammalian cells, has been shown to be important for maintaining the barrier function of lung and kidney epithelia. H. pylori decreases levels of Na,K-ATPase in the plasma membrane of gastric epithelial cells, and the aim of this study was to demonstrate that this reduction led to gastric injury by impairing the epithelial barrier. Similar to H. pylori infection, the inhibition of Na,K-ATPase with ouabain decreased transepithelial electrical resistance and increased paracellular permeability in cell monolayers of human gastric cultured cells, 2D human gastric organoids, and gastric epithelium isolated from gerbils. Similar effects were caused by a partial shRNA silencing of Na,K-ATPase in human gastric organoids. Both H. pylori infection and ouabain exposure disrupted organization of adherens junctions in human gastric epithelia as demonstrated by E-cadherin immunofluorescence. Functional and structural impairment of epithelial integrity with a decrease in Na,K-ATPase amount or activity provides evidence that the H. pylori-induced downregulation of Na,K-ATPase plays a role in the complex mechanism of gastric disease induced by the bacteria.
Collapse
Affiliation(s)
- Olga Vagin
- Department of Pediatrics, DGSOM at UCLA, 10833 LeConte Ave., 12-383 MDCC, Los Angeles, CA 90095, USA; (O.V.); (E.T.)
- VA GLAHS 11301 Wilshire Blvd, Bldg 113, Rm 324, Los Angeles, CA 90073, USA; (M.L.); (J.D.)
| | - Elmira Tokhtaeva
- Department of Pediatrics, DGSOM at UCLA, 10833 LeConte Ave., 12-383 MDCC, Los Angeles, CA 90095, USA; (O.V.); (E.T.)
- VA GLAHS 11301 Wilshire Blvd, Bldg 113, Rm 324, Los Angeles, CA 90073, USA; (M.L.); (J.D.)
| | - Muriel Larauche
- VA GLAHS 11301 Wilshire Blvd, Bldg 113, Rm 324, Los Angeles, CA 90073, USA; (M.L.); (J.D.)
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, DGSOM at UCLA, 650 Charles E Young Dr. S., CHS 43-276, Los Angeles, CA 90095, USA
| | - Joshua Davood
- VA GLAHS 11301 Wilshire Blvd, Bldg 113, Rm 324, Los Angeles, CA 90073, USA; (M.L.); (J.D.)
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, DGSOM at UCLA, 650 Charles E Young Dr. S., CHS 43-276, Los Angeles, CA 90095, USA
| | - Elizabeth A. Marcus
- Department of Pediatrics, DGSOM at UCLA, 10833 LeConte Ave., 12-383 MDCC, Los Angeles, CA 90095, USA; (O.V.); (E.T.)
- VA GLAHS 11301 Wilshire Blvd, Bldg 113, Rm 324, Los Angeles, CA 90073, USA; (M.L.); (J.D.)
| |
Collapse
|
2
|
Contreras RG, Torres-Carrillo A, Flores-Maldonado C, Shoshani L, Ponce A. Na +/K +-ATPase: More than an Electrogenic Pump. Int J Mol Sci 2024; 25:6122. [PMID: 38892309 PMCID: PMC11172918 DOI: 10.3390/ijms25116122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
The sodium pump, or Na+/K+-ATPase (NKA), is an essential enzyme found in the plasma membrane of all animal cells. Its primary role is to transport sodium (Na+) and potassium (K+) ions across the cell membrane, using energy from ATP hydrolysis. This transport creates and maintains an electrochemical gradient, which is crucial for various cellular processes, including cell volume regulation, electrical excitability, and secondary active transport. Although the role of NKA as a pump was discovered and demonstrated several decades ago, it remains the subject of intense research. Current studies aim to delve deeper into several aspects of this molecular entity, such as describing its structure and mode of operation in atomic detail, understanding its molecular and functional diversity, and examining the consequences of its malfunction due to structural alterations. Additionally, researchers are investigating the effects of various substances that amplify or decrease its pumping activity. Beyond its role as a pump, growing evidence indicates that in various cell types, NKA also functions as a receptor for cardiac glycosides like ouabain. This receptor activity triggers the activation of various signaling pathways, producing significant morphological and physiological effects. In this report, we present the results of a comprehensive review of the most outstanding studies of the past five years. We highlight the progress made regarding this new concept of NKA and the various cardiac glycosides that influence it. Furthermore, we emphasize NKA's role in epithelial physiology, particularly its function as a receptor for cardiac glycosides that trigger intracellular signals regulating cell-cell contacts, proliferation, differentiation, and adhesion. We also analyze the role of NKA β-subunits as cell adhesion molecules in glia and epithelial cells.
Collapse
Affiliation(s)
| | | | | | | | - Arturo Ponce
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, Mexico City 07360, Mexico; (R.G.C.); (A.T.-C.); (C.F.-M.); (L.S.)
| |
Collapse
|
3
|
Martínez-Rendón J, Hinojosa L, Xoconostle-Cázares B, Ramírez-Pool JA, Castillo A, Cereijido M, Ponce A. Ouabain Induces Transcript Changes and Activation of RhoA/ROCK Signaling in Cultured Epithelial Cells (MDCK). Curr Issues Mol Biol 2023; 45:7538-7556. [PMID: 37754259 PMCID: PMC10528288 DOI: 10.3390/cimb45090475] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/30/2023] [Accepted: 09/09/2023] [Indexed: 09/28/2023] Open
Abstract
Ouabain, an organic compound with the ability to strengthen the contraction of the heart muscle, was originally derived from plants. It has been observed that certain mammalian species, including humans, naturally produce ouabain, leading to its classification as a new type of hormone. When ouabain binds to Na+/K+-ATPase, it elicits various physiological effects, although these effects are not well characterized. Previous studies have demonstrated that ouabain, within the concentration range found naturally in the body (10 nmol/L), affects the polarity of epithelial cells and their intercellular contacts, such as tight junctions, adherens junctions, and gap junctional communication. This is achieved by activating signaling pathways involving cSrc and Erk1/2. To further investigate the effects of ouabain within the hormonally relevant concentration range (10 nmol/L), mRNA-seq, a high-throughput sequencing technique, was employed to identify differentially expressed transcripts. The discovery that the transcript encoding MYO9A was among the genes affected prompted an exploration of whether RhoA and its downstream effector ROCK were involved in the signaling pathways through which ouabain influences cell-to-cell contacts in epithelial cells. Supporting this hypothesis, this study reveals the following: (1) Ouabain increases the activation of RhoA. (2) Treatment with inhibitors of RhoA activation (Y27) and ROCK (C3) eliminates the enhancing effect of ouabain on the tight junction seal and intercellular communication via gap junctions. These findings further support the notion that ouabain acts as a hormone to emphasize the epithelial phenotype.
Collapse
Affiliation(s)
- Jacqueline Martínez-Rendón
- Department of Physiology, Biophysics and Neurosciences, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico 07360, Mexico; (J.M.-R.); (L.H.); (A.C.); (M.C.)
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S., Campus UAZ Siglo XXI-L1, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico
| | - Lorena Hinojosa
- Department of Physiology, Biophysics and Neurosciences, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico 07360, Mexico; (J.M.-R.); (L.H.); (A.C.); (M.C.)
| | - Beatriz Xoconostle-Cázares
- Department of Biotechnology and Bioengineering, CINVESTAV-IPN, Ciudad de Mexico 07360, Mexico; (B.X.-C.); (J.A.R.-P.)
| | - José Abrahán Ramírez-Pool
- Department of Biotechnology and Bioengineering, CINVESTAV-IPN, Ciudad de Mexico 07360, Mexico; (B.X.-C.); (J.A.R.-P.)
| | - Aída Castillo
- Department of Physiology, Biophysics and Neurosciences, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico 07360, Mexico; (J.M.-R.); (L.H.); (A.C.); (M.C.)
| | - Marcelino Cereijido
- Department of Physiology, Biophysics and Neurosciences, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico 07360, Mexico; (J.M.-R.); (L.H.); (A.C.); (M.C.)
| | - Arturo Ponce
- Department of Physiology, Biophysics and Neurosciences, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico 07360, Mexico; (J.M.-R.); (L.H.); (A.C.); (M.C.)
| |
Collapse
|
4
|
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
|
5
|
In Vitro Cytotoxicity Induced by the Bufadienolides 1α,2α-Epoxyscillirosidine and Lanceotoxin B on Rat Myocardial and Mouse Neuroblastoma Cell Lines. Toxins (Basel) 2019; 11:toxins11010014. [PMID: 30609690 PMCID: PMC6356634 DOI: 10.3390/toxins11010014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 12/21/2018] [Accepted: 12/23/2018] [Indexed: 11/22/2022] Open
Abstract
Consumption of bufadienolide-containing plants are responsible for many livestock mortalities annually. Bufadienolides are divided into two groups; non-cumulative bufadienolides and cumulative bufadienolides. Cumulative bufadienolides are referred to as neurotoxic, as the chronic intoxication with this type of bufadienolide results in a paretic/paralytic syndrome known as ‘krimpsiekte’. The in vitro cytotoxicity of a non-cumulative bufadienolide, 1α,2α-epoxyscillirosidine, and a cumulative bufadienolide, lanceotoxin B, were compared using the MTT ((3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide reduction) assay after exposing rat myocardial (H9c2) and mouse neuroblastoma (Neuro-2a) cell lines. The effect of these two bufadienolides on cell ultrastructure was also investigated using transmission electron microscopy (TEM). H9c2 cells exhibited greater cytotoxicity when exposed to 1α,2α-epoxyscillirosidine, compared to lanceotoxin B. In contrast, Neuro-2a cells were more susceptible to lanceotoxin B. The EC50 (half maximal effective concentration) of lanceotoxin B exposure of Neuro-2a cells for 24–72 h ranged from 4.4–5.5 µM compared to EC50s of 35.7–37.6 µM for 1α,2α-epoxyscillirosidine exposure of Neuro-2a cells over the same period. 1α,2α-Epoxyscillirosidine induced extensive vacuolization in both cell types, with swollen RER (rough endoplasmic reticulum) and perinuclear spaces. Lanceotoxin B caused swelling of the mitochondria and sequestration of cytoplasmic material within autophagic vesicles. These results corroborate the notion that cumulative bufadienolides are neurotoxic.
Collapse
|
6
|
Orellana AM, Leite JA, Kinoshita PF, Vasconcelos AR, Andreotti DZ, de Sá Lima L, Xavier GF, Kawamoto EM, Scavone C. Ouabain increases neuronal branching in hippocampus and improves spatial memory. Neuropharmacology 2018; 140:260-274. [PMID: 30099050 DOI: 10.1016/j.neuropharm.2018.08.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 07/05/2018] [Accepted: 08/06/2018] [Indexed: 12/20/2022]
Abstract
Previous research shows Ouabain (OUA) to bind Na, K-ATPase, thereby triggering a number of signaling pathways, including the transcription factors NFᴋB and CREB. These transcription factors play a key role in the regulation of BDNF and WNT-β-catenin signaling cascades, which are involved in neuroprotection and memory regulation. This study investigated the effects of OUA (10 nM) in the modulation of the principal signaling pathways involved in morphological plasticity and memory formation in the hippocampus of adult rats. The results show intrahippocampal injection of OUA 10 nM to activate the Wnt/β-Catenin signaling pathway and to increase CREB/BDNF and NFᴋB levels. These effects contribute to important changes in the cellular microenvironment, resulting in enhanced levels of dendritic branching in hippocampal neurons, in association with an improvement in spatial reference memory and the inhibition of long-term memory extinction.
Collapse
Affiliation(s)
- Ana Maria Orellana
- Department of Pharmacology, Institute of Biomedical Science I University of São Paulo, Room 338, Av. Prof. Lineu Prestes, 1524, ICB I, Cidade Universitária, 05508-900, São Paulo, SP. Brazil.
| | - Jacqueline Alves Leite
- Department of Pharmacology, Institute of Biomedical Science I University of São Paulo, Room 338, Av. Prof. Lineu Prestes, 1524, ICB I, Cidade Universitária, 05508-900, São Paulo, SP. Brazil.
| | - Paula Fernanda Kinoshita
- Department of Pharmacology, Institute of Biomedical Science I University of São Paulo, Room 338, Av. Prof. Lineu Prestes, 1524, ICB I, Cidade Universitária, 05508-900, São Paulo, SP. Brazil.
| | - Andrea Rodrigues Vasconcelos
- Department of Pharmacology, Institute of Biomedical Science I University of São Paulo, Room 338, Av. Prof. Lineu Prestes, 1524, ICB I, Cidade Universitária, 05508-900, São Paulo, SP. Brazil.
| | - Diana Zukas Andreotti
- Department of Pharmacology, Institute of Biomedical Science I University of São Paulo, Room 338, Av. Prof. Lineu Prestes, 1524, ICB I, Cidade Universitária, 05508-900, São Paulo, SP. Brazil.
| | - Larissa de Sá Lima
- Department of Pharmacology, Institute of Biomedical Science I University of São Paulo, Room 338, Av. Prof. Lineu Prestes, 1524, ICB I, Cidade Universitária, 05508-900, São Paulo, SP. Brazil.
| | - Gilberto Fernando Xavier
- Department of Physiology, Institute of Bioscience, University of São Paulo, Adress: Rua do Matão, Travessa 14, 101, São Paulo, 05508-090, Brazil.
| | - Elisa Mitiko Kawamoto
- Department of Pharmacology, Institute of Biomedical Science I University of São Paulo, Room 338, Av. Prof. Lineu Prestes, 1524, ICB I, Cidade Universitária, 05508-900, São Paulo, SP. Brazil.
| | - Cristoforo Scavone
- Department of Pharmacology, Institute of Biomedical Science I University of São Paulo, Room 338, Av. Prof. Lineu Prestes, 1524, ICB I, Cidade Universitária, 05508-900, São Paulo, SP. Brazil.
| |
Collapse
|
7
|
Taub M. Gene Level Regulation of Na,K-ATPase in the Renal Proximal Tubule Is Controlled by Two Independent but Interacting Regulatory Mechanisms Involving Salt Inducible Kinase 1 and CREB-Regulated Transcriptional Coactivators. Int J Mol Sci 2018; 19:E2086. [PMID: 30021947 PMCID: PMC6073390 DOI: 10.3390/ijms19072086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 12/26/2022] Open
Abstract
For many years, studies concerning the regulation of Na,K-ATPase were restricted to acute regulatory mechanisms, which affected the phosphorylation of Na,K-ATPase, and thus its retention on the plasma membrane. However, in recent years, this focus has changed. Na,K-ATPase has been established as a signal transducer, which becomes part of a signaling complex as a consequence of ouabain binding. Na,K-ATPase within this signaling complex is localized in caveolae, where Na,K-ATPase has also been observed to regulate Inositol 1,4,5-Trisphosphate Receptor (IP3R)-mediated calcium release. This latter association has been implicated as playing a role in signaling by G Protein Coupled Receptors (GPCRs). Here, the consequences of signaling by renal effectors that act via such GPCRs are reviewed, including their regulatory effects on Na,K-ATPase gene expression in the renal proximal tubule (RPT). Two major types of gene regulation entail signaling by Salt Inducible Kinase 1 (SIK1). On one hand, SIK1 acts so as to block signaling via cAMP Response Element (CRE) Binding Protein (CREB) Regulated Transcriptional Coactivators (CRTCs) and on the other hand, SIK1 acts so as to stimulate signaling via the Myocyte Enhancer Factor 2 (MEF2)/nuclear factor of activated T cell (NFAT) regulated genes. Ultimate consequences of these pathways include regulatory effects which alter the rate of transcription of the Na,K-ATPase β1 subunit gene atp1b1 by CREB, as well as by MEF2/NFAT.
Collapse
Affiliation(s)
- Mary Taub
- Biochemistry Dept., Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 955 Main Street, Suite 4902, Buffalo, NY 14203, USA.
| |
Collapse
|
8
|
On the Many Actions of Ouabain: Pro-Cystogenic Effects in Autosomal Dominant Polycystic Kidney Disease. Molecules 2017; 22:molecules22050729. [PMID: 28467389 PMCID: PMC5688955 DOI: 10.3390/molecules22050729] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/19/2017] [Accepted: 04/30/2017] [Indexed: 02/07/2023] Open
Abstract
Ouabain and other cardenolides are steroidal compounds originally discovered in plants. Cardenolides were first used as poisons, but after finding their beneficial cardiotonic effects, they were rapidly included in the medical pharmacopeia. The use of cardenolides to treat congestive heart failure remained empirical for centuries and only relatively recently, their mechanisms of action became better understood. A breakthrough came with the discovery that ouabain and other cardenolides exist as endogenous compounds that circulate in the bloodstream of mammals. This elevated these compounds to the category of hormones and opened new lines of investigation directed to further study their biological role. Another important discovery was the finding that the effect of ouabain was mediated not only by inhibition of the activity of the Na,K-ATPase (NKA), but by the unexpected role of NKA as a receptor and a signal transducer, which activates a complex cascade of intracellular second messengers in the cell. This broadened the interest for ouabain and showed that it exerts actions that go beyond its cardiotonic effect. It is now clear that ouabain regulates multiple cell functions, including cell proliferation and hypertrophy, apoptosis, cell adhesion, cell migration, and cell metabolism in a cell and tissue type specific manner. This review article focuses on the cardenolide ouabain and discusses its various in vitro and in vivo effects, its role as an endogenous compound, its mechanisms of action, and its potential use as a therapeutic agent; placing especial emphasis on our findings of ouabain as a pro-cystogenic agent in autosomal dominant polycystic kidney disease (ADPKD).
Collapse
|
9
|
Na⁺ i,K⁺ i-Dependent and -Independent Signaling Triggered by Cardiotonic Steroids: Facts and Artifacts. Molecules 2017; 22:molecules22040635. [PMID: 28420099 PMCID: PMC6153942 DOI: 10.3390/molecules22040635] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 03/31/2017] [Accepted: 04/11/2017] [Indexed: 11/17/2022] Open
Abstract
Na⁺,K⁺-ATPase is the only known receptor of cardiotonic steroids (CTS) whose interaction with catalytic α-subunits leads to inhibition of this enzyme. As predicted, CTS affect numerous cellular functions related to the maintenance of the transmembrane gradient of monovalent cations, such as electrical membrane potential, cell volume, transepithelial movement of salt and osmotically-obliged water, symport of Na⁺ with inorganic phosphate, glucose, amino acids, nucleotides, etc. During the last two decades, it was shown that side-by-side with these canonical Na⁺i/K⁺i-dependent cellular responses, long-term exposure to CTS affects transcription, translation, tight junction, cell adhesion and exhibits tissue-specific impact on cell survival and death. It was also shown that CTS trigger diverse signaling cascades via conformational transitions of the Na⁺,K⁺-ATPase α-subunit that, in turn, results in the activation of membrane-associated non-receptor tyrosine kinase Src, phosphatidylinositol 3-kinase and the inositol 1,4,5-triphosphate receptor. These findings allowed researchers to propose that endogenous CTS might be considered as a novel class of steroid hormones. We focus our review on the analysis of the relative impact Na⁺i,K⁺i-mediated and -independent pathways in cellular responses evoked by CTS.
Collapse
|
10
|
Klimanova EA, Tverskoi AM, Koltsova SV, Sidorenko SV, Lopina OD, Tremblay J, Hamet P, Kapilevich LV, Orlov SN. Time- and dose dependent actions of cardiotonic steroids on transcriptome and intracellular content of Na + and K +: a comparative analysis. Sci Rep 2017; 7:45403. [PMID: 28345607 PMCID: PMC5366943 DOI: 10.1038/srep45403] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 02/17/2017] [Indexed: 01/07/2023] Open
Abstract
Recent studies demonstrated that in addition to Na+,K+-ATPase inhibition cardiotonic steroids (CTSs) affect diverse intracellular signaling pathways. This study examines the relative impact of [Na+]i/[K+]i-mediated and -independent signaling in transcriptomic changes triggered by the endogenous CTSs ouabain and marinobufagenin (MBG) in human umbilical vein endothelial cells (HUVEC). We noted that prolongation of incubation increased the apparent affinity for ouabain estimated by the loss of [K+]i and gain of [Na+]i. Six hour exposure of HUVEC to 100 and 3,000 nM ouabain resulted in elevation of the [Na+]i/[K+]i ratio by ~15 and 80-fold and differential expression of 258 and 2185 transcripts, respectively. Neither [Na+]i/[K+]i ratio nor transcriptome were affected by 6-h incubation with 30 nM ouabain. The 96-h incubation with 3 nM ouabain or 30 nM MBG elevated the [Na+]i/[K+]i ratio by ~14 and 3-fold and led to differential expression of 880 and 484 transcripts, respectively. These parameters were not changed after 96-h incubation with 1 nM ouabain or 10 nM MBG. Thus, our results demonstrate that elevation of the [Na+]i/[K+]i ratio is an obligatory step for transcriptomic changes evoked by CTS in HUVEC. The molecular origin of upstream [Na+]i/[K+]i sensors involved in transcription regulation should be identified in forthcoming studies.
Collapse
Affiliation(s)
| | - Artem M Tverskoi
- Faculty of Biology, M. V. Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Svetlana V Koltsova
- Faculty of Biology, M. V. Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Svetlana V Sidorenko
- Faculty of Biology, M. V. Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Olga D Lopina
- Faculty of Biology, M. V. Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Johanne Tremblay
- Research Centre, University of Montreal Hospital (CRCHUM), Montreal, H2X 0A9, Canada
| | - Pavel Hamet
- Research Centre, University of Montreal Hospital (CRCHUM), Montreal, H2X 0A9, Canada
| | | | - Sergei N Orlov
- Faculty of Biology, M. V. Lomonosov Moscow State University, Moscow, 119234, Russia.,National Research Tomsk State University, Tomsk, 634050, Russia
| |
Collapse
|
11
|
New Role for FDA-Approved Drugs in Combating Antibiotic-Resistant Bacteria. Antimicrob Agents Chemother 2016; 60:3717-29. [PMID: 27067323 DOI: 10.1128/aac.00326-16] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/31/2016] [Indexed: 12/16/2022] Open
Abstract
Antibiotic resistance in medically relevant bacterial pathogens, coupled with a paucity of novel antimicrobial discoveries, represents a pressing global crisis. Traditional drug discovery is an inefficient and costly process; however, systematic screening of Food and Drug Administration (FDA)-approved therapeutics for other indications in humans offers a rapid alternative approach. In this study, we screened a library of 780 FDA-approved drugs to identify molecules that rendered RAW 264.7 murine macrophages resistant to cytotoxicity induced by the highly virulent Yersinia pestis CO92 strain. Of these compounds, we identified 94 not classified as antibiotics as being effective at preventing Y. pestis-induced cytotoxicity. A total of 17 prioritized drugs, based on efficacy in in vitro screens, were chosen for further evaluation in a murine model of pneumonic plague to delineate if in vitro efficacy could be translated in vivo Three drugs, doxapram (DXP), amoxapine (AXPN), and trifluoperazine (TFP), increased animal survivability despite not exhibiting any direct bacteriostatic or bactericidal effect on Y. pestis and having no modulating effect on crucial Y. pestis virulence factors. These findings suggested that DXP, AXPN, and TFP may modulate host cell pathways necessary for disease pathogenesis. Finally, to further assess the broad applicability of drugs identified from in vitro screens, the therapeutic potential of TFP, the most efficacious drug in vivo, was evaluated in murine models of Salmonella enterica serovar Typhimurium and Clostridium difficile infections. In both models, TFP treatment resulted in increased survivability of infected animals. Taken together, these results demonstrate the broad applicability and potential use of nonantibiotic FDA-approved drugs to combat respiratory and gastrointestinal bacterial pathogens.
Collapse
|
12
|
Critical role of the α1-Na(+), K(+)-ATPase subunit in insensitivity of rodent cells to cytotoxic action of ouabain. Apoptosis 2016; 20:1200-10. [PMID: 26067145 DOI: 10.1007/s10495-015-1144-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In rodents, ubiquitous α1-Na(+), K(+)-ATPase is inhibited by ouabain and other cardiotonic steroids (CTS) at ~10(3)-fold higher concentrations than those effective in other mammals. To examine the specific roles of the CTS-sensitive α1S- and CTS-resistant α1R-Na(+), K(+)-ATPase isoforms, we compared the effects of ouabain on intracellular Na(+) and K(+) content, cell survival, and mitogen-activated protein kinases (MAPK) in human and rat vascular smooth muscle cells (HASMC and RASMC), human and rat endothelial cells (HUVEC and RAEC), and human and rat brain astrocytes. 6-h exposure of HASMC and HUVEC to 3 μM ouabain dramatically increased the intracellular [Na(+)]/[K(+)] ratio to the same extend as in RASMC and RAEC treated with 3000 μM ouabain. In 24, 3 μM ouabain triggered the death of all types of human cells used in this study. Unlike human cells, we did not detect any effect of 3000-5000 μM ouabain on the survival of rat cells, or smooth muscle cells from mouse aorta (MASMC). Unlike in the wild-type α1(R/R) mouse, ouabain triggered death of MASMC from α1(S/S) mouse expressing human α1-Na(+), K(+)-ATPase. Furthermore, transfection of HUVEC with rat α1R-Na(+), K(+)-ATPase protected them from the ouabain-induced death. In HUVEC, ouabain led to phosphorylation of p38 MAPK, whereas in RAEC it stimulated phosphorylation of ERK1/2. Overall, our results, demonstrate that the drastic differences in cytotoxic action of ouabain on human and rodent cells are caused by unique features of α1S/α1R-Na(+), K(+)-ATPase, rather than by any downstream CTS-sensitive/resistant components of the cell death machinery.
Collapse
|
13
|
Khundmiri SJ. Advances in understanding the role of cardiac glycosides in control of sodium transport in renal tubules. J Endocrinol 2014; 222:R11-24. [PMID: 24781255 DOI: 10.1530/joe-13-0613] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cardiotonic steroids have been used for the past 200 years in the treatment of congestive heart failure. As specific inhibitors of membrane-bound Na(+)/K(+) ATPase, they enhance cardiac contractility through increasing myocardial cell calcium concentration in response to the resulting increase in intracellular Na concentration. The half-minimal concentrations of cardiotonic steroids required to inhibit Na(+)/K(+) ATPase range from nanomolar to micromolar concentrations. In contrast, the circulating levels of cardiotonic steroids under physiological conditions are in the low picomolar concentration range in healthy subjects, increasing to high picomolar levels under pathophysiological conditions including chronic kidney disease and heart failure. Little is known about the physiological function of low picomolar concentrations of cardiotonic steroids. Recent studies have indicated that physiological concentrations of cardiotonic steroids acutely stimulate the activity of Na(+)/K(+) ATPase and activate an intracellular signaling pathway that regulates a variety of intracellular functions including cell growth and hypertrophy. The effects of circulating cardiotonic steroids on renal salt handling and total body sodium homeostasis are unknown. This review will focus on the role of low picomolar concentrations of cardiotonic steroids in renal Na(+)/K(+) ATPase activity, cell signaling, and blood pressure regulation.
Collapse
Affiliation(s)
- Syed Jalal Khundmiri
- Division of Nephrology and HypertensionDepartment of MedicineDepartment of Physiology and BiophysicsUniversity of Louisville, 570 S. Preston Street, Louisville, Kentucky 40202, USADivision of Nephrology and HypertensionDepartment of MedicineDepartment of Physiology and BiophysicsUniversity of Louisville, 570 S. Preston Street, Louisville, Kentucky 40202, USA
| |
Collapse
|
14
|
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]
|
15
|
de Souza WF, Barbosa LA, Liu L, de Araujo WM, de-Freitas-Junior JCM, Fortunato-Miranda N, Fontes CFL, Morgado-Díaz JA. Ouabain-induced alterations of the apical junctional complex involve α1 and β1 Na,K-ATPase downregulation and ERK1/2 activation independent of caveolae in colorectal cancer cells. J Membr Biol 2013; 247:23-33. [PMID: 24186357 DOI: 10.1007/s00232-013-9607-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 10/14/2013] [Indexed: 12/22/2022]
Abstract
Studies have reported that Na,K-ATPase interacts with E-cadherin to stabilize (AJs) and regulate the expression of claudins, the main proteins present in the tight junction (TJ) in epithelial cells containing caveolae. However, the role of this ATPase in the regulation of the AJ and TJ proteins in colorectal cancer cells as well as the molecular events underlying this event in a caveolae-independent system remain undefined. In the present study, we used ouabain, a classic drug known to inhibit Na,K-ATPase, and Caco-2 cells, which are a well-established human colorectal cancer model that does not exhibit caveolae. We demonstrated that ouabain treatment resulted in a reduction of the β1 Na,K-ATPase protein and cell redistribution of the AJ proteins E-cadherin and β-catenin, as well as the α1 Na,K-ATPase subunit. Furthermore, ouabain increased claudin-3 protein levels, impaired the TJ barrier function and increased cell viability and proliferation during the early stages of treatment. Additionally, the observed ouabain-induced events were dependent on the activation of ERK1/2 signaling; but in contrast to previous studies, this signaling cascade was caveolae-independent. In conclusion, our findings strongly suggest that α1 and β1 Na,K-ATPase downregulation and ERK1/2 activation induced by ouabain are interlinked events that play an important role during cell-cell adhesion loss, which is an important step during the tumor progression of colorectal carcinomas.
Collapse
Affiliation(s)
- Waldemir Fernandes de Souza
- Programa de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 5° andar, Rio de Janeiro, RJ, 20231-050, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
16
|
de Souza WF, Fortunato-Miranda N, Robbs BK, de Araujo WM, de-Freitas-Junior JC, Bastos LG, Viola JPB, Morgado-Díaz JA. Claudin-3 overexpression increases the malignant potential of colorectal cancer cells: roles of ERK1/2 and PI3K-Akt as modulators of EGFR signaling. PLoS One 2013; 8:e74994. [PMID: 24069372 PMCID: PMC3777902 DOI: 10.1371/journal.pone.0074994] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 08/09/2013] [Indexed: 12/19/2022] Open
Abstract
The altered expressions of claudin proteins have been reported during the tumorigenesis of colorectal cancer. However, the molecular mechanisms that regulate these events in this cancer type are poorly understood. Here, we report that epidermal growth factor (EGF) increases the expression of claudin-3 in human colorectal adenocarcinoma HT-29 cells. This increase was related to increased cell migration and the formation of anchorage-dependent and anchorage-independent colonies. We further showed that the ERK1/2 and PI3K-Akt pathways were involved in the regulation of these effects because specific pharmacological inhibition blocked these events. Genetic manipulation of claudin-1 and claudin-3 in HT-29 cells showed that the overexpression of claudin-1 resulted in decreased cell migration; however, migration was not altered in cells that overexpressed claudin-3. Furthermore, the overexpression of claudin-3, but not that of claudin-1, increased the tight junction-related paracellular flux of macromolecules. Additionally, an increased formation of anchorage-dependent and anchorage-independent colonies were observed in cells that overexpressed claudin-3, while no such changes were observed when claudin-1 was overexpressed. Finally, claudin-3 silencing alone despite induce increase proliferation, and the formation of anchoragedependent and -independent colonies, it was able to prevent the EGF-induced increased malignant potential. In conclusion, our results show a novel role for claudin-3 overexpression in promoting the malignant potential of colorectal cancer cells, which is potentially regulated by the EGF-activated ERK1/2 and PI3K-Akt pathways.
Collapse
Affiliation(s)
- Waldemir F. de Souza
- Grupo de Biologia Estrutural, Programa de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Natalia Fortunato-Miranda
- Grupo de Biologia Estrutural, Programa de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Bruno K. Robbs
- Grupo de Regulação Gênica, Programa de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Wallace M. de Araujo
- Grupo de Biologia Estrutural, Programa de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Julio C. de-Freitas-Junior
- Grupo de Biologia Estrutural, Programa de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Lilian G. Bastos
- Grupo de Biologia Estrutural, Programa de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - João P. B. Viola
- Grupo de Regulação Gênica, Programa de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - José A. Morgado-Díaz
- Grupo de Biologia Estrutural, Programa de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
17
|
Blanco G, Wallace DP. Novel role of ouabain as a cystogenic factor in autosomal dominant polycystic kidney disease. Am J Physiol Renal Physiol 2013; 305:F797-812. [PMID: 23761677 DOI: 10.1152/ajprenal.00248.2013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The classic role of the Na-K-ATPase is that of a primary active transporter that utilizes cell energy to establish and maintain transmembrane Na(+) and K(+) gradients to preserve cell osmotic stability, support cell excitability, and drive secondary active transport. Recent studies have revealed that Na-K-ATPase located within cholesterol-containing lipid rafts serves as a receptor for cardiotonic steroids, including ouabain. Traditionally, ouabain was viewed as a toxin produced only in plants, and it was used in relatively high concentrations to experimentally block the pumping action of the Na-K-ATPase. However, the new and unexpected role of the Na-K-ATPase as a signal transducer revealed a novel facet for ouabain in the regulation of a myriad of cell functions, including cell proliferation, hypertrophy, apoptosis, mobility, and metabolism. The seminal discovery that ouabain is endogenously produced in mammals and circulates in plasma has fueled the interest in this endogenous molecule as a potentially important hormone in normal physiology and disease. In this article, we review the role of the Na-K-ATPase as an ion transporter in the kidney, the experimental evidence for ouabain as a circulating hormone, the function of the Na-K-ATPase as a signal transducer that mediates ouabain's effects, and novel results for ouabain-induced Na-K-ATPase signaling in cystogenesis of autosomal dominant polycystic kidney disease.
Collapse
Affiliation(s)
- Gustavo Blanco
- Dept. of Molecular and Integrative Physiology, 3901 Rainbow Blvd., Kansas City, KS 66160.
| | | |
Collapse
|
18
|
Orlov SN, Platonova AA, Hamet P, Grygorczyk R. Cell volume and monovalent ion transporters: their role in cell death machinery triggering and progression. Am J Physiol Cell Physiol 2013; 305:C361-72. [PMID: 23615964 DOI: 10.1152/ajpcell.00040.2013] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cell death is accompanied by the dissipation of electrochemical gradients of monovalent ions across the plasma membrane that, in turn, affects cell volume via modulation of intracellular osmolyte content. In numerous cell types, apoptotic and necrotic stimuli caused cell shrinkage and swelling, respectively. Thermodynamics predicts a cell type-specific rather than an ubiquitous impact of monovalent ion transporters on volume perturbations in dying cells, suggesting their diverse roles in the cell death machinery. Indeed, recent data showed that apoptotic collapse may occur in the absence of cell volume changes and even follow cell swelling rather than shrinkage. Moreover, side-by-side with cell volume adjustment, monovalent ion transporters contribute to cell death machinery engagement independently of volume regulation via cell type-specific signaling pathways. Thus, inhibition of Na(+)-K(+)-ATPase by cardiotonic steroids (CTS) rescues rat vascular smooth muscle cells from apoptosis via a novel Na(+)i-K(+)i-mediated, Ca(2+)i-independent mechanism of excitation-transcription coupling. In contrast, CTS kill renal epithelial cells independently of Na(+)-K(+)-ATPase inhibition and increased [Na(+)]i/[K(+)]i ratio. The molecular origin of [Na(+)]i/[K(+)]i sensors involved in the inhibition of apoptosis as well as upstream intermediates of Na(+)i/K(+)i-independent death signaling triggered by CTS remain unknown.
Collapse
Affiliation(s)
- Sergei N Orlov
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.
| | | | | | | |
Collapse
|
19
|
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
|
20
|
Elbaz HA, Stueckle TA, Tse W, Rojanasakul Y, Dinu CZ. Digitoxin and its analogs as novel cancer therapeutics. Exp Hematol Oncol 2012; 1:4. [PMID: 23210930 PMCID: PMC3506989 DOI: 10.1186/2162-3619-1-4] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 04/05/2012] [Indexed: 01/18/2023] Open
Abstract
A growing body of evidence indicates that digitoxin cardiac glycoside is a promising anticancer agent when used at therapeutic concentrations. Digitoxin has a prolonged half-life and a well-established clinical profile. New scientific avenues have shown that manipulating the chemical structure of the saccharide moiety of digitoxin leads to synthetic analogs with increased cytotoxic activity. However, the anticancer mechanism of digitoxin or synthetic analogs is still subject to study while concerns about digitoxin's cardiotoxicity preclude its clinical application in cancer therapeutics. This review focuses on digitoxin and its analogs, and their cytotoxicity against cancer cells. Moreover, a new perspective on the pharmacological aspects of digitoxin and its analogs is provided to emphasize new research directions for developing potent chemotherapeutic drugs.
Collapse
Affiliation(s)
- Hosam A Elbaz
- Department of Basic Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA.
| | | | | | | | | |
Collapse
|
21
|
Weidemann H. "The Lower Threshold" phenomenon in tumor cells toward endogenous digitalis-like compounds: Responsible for tumorigenesis? J Carcinog 2012; 11:2. [PMID: 22438768 PMCID: PMC3307333 DOI: 10.4103/1477-3163.92999] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 12/01/2011] [Indexed: 02/06/2023] Open
Abstract
Since their first discovery as potential anti-cancer drugs decades ago, there is increasing evidence that digitalis-like compounds (DLC) have anti-tumor effects. Less is known about endogenous DLC (EDLC) metabolism and regulation. As stress hormones synthesized in and secreted from the adrenal gland, they likely take part in the hypothalamo-pituitary-adrenal (HPA) axis. In a previous study, we revealed reduced EDLC concentrations in plasma and organs from immune-compromised animals and proposed that a similar situation of a deregulated HPA axis with "adrenal EDLF exhaustion" may contribute to tumorigenesis in chronic stress situations. Here, we put forward the hypothesis that a lowered EDLC response threshold of tumor cells as compared with normal cells increases the risk of tumorigenesis, especially in those individuals with reduced EDLC plasma concentrations after chronic stress exposure. We will evaluate this hypothesis by (a) summarizing the effects of different DLC concentrations on tumor as compared with normal cells and (b) reviewing some essential differences in the Na/K-ATPase of tumor as compared with normal cells (isoform pattern, pump activity, mutations of other signalosome receptors). We will conclude that (1) tumor cells, indeed, seem to have their individual "physiologic" EDLC response range that already starts at pmolar levels and (2) that individuals with markedly reduced (pmolar) EDLC plasma levels are predisposed to cancer because these EDLC concentrations will predominantly stimulate the proliferation of tumor cells. Finally, we will summarize preliminary results from our department supporting this hypothesis.
Collapse
Affiliation(s)
- Heidrun Weidemann
- Department of Oncology, Hadassah-Hebrew University, Medical Center, Jerusalem, Israel
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
Ferrandi M, Molinari I, Torielli L, Padoani G, Salardi S, Rastaldi MP, Ferrari P, Bianchi G. Adducin- and ouabain-related gene variants predict the antihypertensive activity of rostafuroxin, part 1: experimental studies. Sci Transl Med 2011; 2:59ra86. [PMID: 21106940 DOI: 10.1126/scitranslmed.3001815] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Essential hypertension is a complex, multifactorial disease associated with a high cardiovascular risk and whose genetic-molecular basis is heterogeneous and largely unknown. Although multiple antihypertensive therapies are available, the large individual variability in drug response results in only a modest reduction of the cardiovascular risk and unsatisfactory control of blood pressure in the hypertensive population as a whole. Two mechanisms, among others, are associated with essential hypertension and related organ damage: mutant α-adducin variants and high concentrations of endogenous ouabain. An antihypertensive agent, rostafuroxin, selectively inhibits these mechanisms in rodents. We investigated the molecular and functional effects of mutant α-adducin, ouabain, and rostafuroxin in hypertensive rats, human cells, and cell-free systems and demonstrated that both mutant α-adducin variants and the ouabain-Na,K-ATPase (Na(+)- and K(+)-dependent adenosine triphosphatase) complex can interact with the Src-SH2 (Src homology 2) domain, increasing Src activity and the Src-dependent Na,K-ATPase phosphorylation and activity. Wild-type α-adducin or Na,K-ATPase in the absence of ouabain showed no interaction with the Src-SH2 domain. Rostafuroxin disrupted the interactions between the Src-SH2 domain and mutant α-adducin or the ouabain-Na,K-ATPase complex and blunted Src activation and Na,K-ATPase phosphorylation, resulting in blood pressure normalization in the hypertensive rats. We have also shown the translatability of these data to humans in a pharmacogenomic clinical trial, as described in the companion paper.
Collapse
Affiliation(s)
- Mara Ferrandi
- Prassis sigma-tau Research Institute, Settimo Milanese, Milan 20019, Italy
| | | | | | | | | | | | | | | |
Collapse
|
25
|
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
|
26
|
Platonova A, Koltsova S, Maksimov GV, Grygorczyk R, Orlov SN. The death of ouabain-treated renal epithelial C11-MDCK cells is not mediated by swelling-induced plasma membrane rupture. J Membr Biol 2011; 241:145-54. [PMID: 21584679 DOI: 10.1007/s00232-011-9371-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 05/05/2011] [Indexed: 10/18/2022]
Abstract
This study examined the role of cell volume modulation in plasma membrane rupture and death documented in ouabain-treated renal epithelial cells. Long-term exposure to ouabain caused massive death of C11-MDCK (Madin-Darby canine kidney) epithelial cells, documented by their detachment, chromatin cleavage and complete loss of lactate dehydrogenase (LDH), but did not affect the survival of vascular smooth muscle cells (VSMCs) from the rat aorta. Unlike the distinct impact on cell survival, 2-h exposure to ouabain led to sharp elevation of the [Na⁺](i)/[K⁺](i) ratio in both cell types. A similar increment of Na⁺(i) content was evoked by sustained inhibition of Na⁺,K⁺-ATPase in K⁺-free medium. However, in contrast to ouabain, C11-MDCK cells survived perfectly during 24-h exposure to K⁺-free medium. At 3 h, the volume of ouabain-treated C11-MDCK cells and VSMCs, measured by the recently developed dual-image surface reconstruction technique, was increased by 16 and 12%, respectively, whereas 5-10 min before the detachment of ouabain-treated C11-MDCK cells, their volume was augmented by ~30-40%. To examine the role of modest swelling in the plasma membrane rupture of ouabain-treated cells, we compared actions of hypotonic medium on volume and LDH release. We observed that LDH release from hypoosmotically swollen C11-MDCK cells was triggered when their volume was increased by approximately fivefold. Thus, our results showed that the rupture of plasma membranes in ouabain-treated C11-MDCK cells was not directly caused by cell volume modulation evoked by Na⁺,K⁺-ATPase inhibition and inversion of the [Na⁺](i)/[K⁺](i) ratio.
Collapse
Affiliation(s)
- Alexandra Platonova
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM)-Technopôle Angus, 2901 Rachel Est, Montreal, QC H1W4A4, Canada
| | | | | | | | | |
Collapse
|
27
|
Akimova OA, Lopina OD, Rubtsov AM, Hamet P, Orlov SN. Investigation of mechanism of p38 MAPK activation in renal epithelial cell from distal tubules triggered by cardiotonic steroids. BIOCHEMISTRY (MOSCOW) 2011; 75:971-8. [PMID: 21073417 DOI: 10.1134/s0006297910080043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Ouabain and other cardiotonic steroids (CTS) kill renal epithelial cells from distal tubules (C7-MDCK) via interaction with Na,K-ATPase but independently of inhibition of Na,K-ATPase-mediated ion fluxes. Recently, we demonstrated that modest intracellular acidification and inhibition of p38 MAPK suppress death of C7-MDCK cells triggered by ouabain. In the present study we investigate the mechanism of p38 MAPK activation in renal epithelial cell from distal tubules evoked by cardiotonic steroids. Using Na+/K+ ionophores (monensin, nigericin) and media with different content of monovalent cations, we revealed that p38 MAPK phosphorylation in ouabain-treated renal epithelial cells is not caused by Na,K-ATPase inhibition and inversion of the [Na+](i)/[K+](i) ratio. We also demonstrated that attenuation of pH from 7.45 to 6.75 did not alter the level of p38 MAPK phosphorylation observed in ouabain-treated cells. Inhibitors of PKA, PKC, and PKG as well as protein phosphatases were unable to abolish p38 MAPK activation triggered by ouabain. Using phosphotyrosine antibodies we did not detect any effect of ouabain on activation of tyrosine kinases. Thus, our results show that activation of p38 MAPK and cytotoxic action of CTS are independent of intracellular Na+, K+, and H+ concentrations. The molecular origin of intermediates of death signaling induced by CTS via conformation changes of Na,K-ATPase with following activation of p38 MAPK should be examined further.
Collapse
Affiliation(s)
- O A Akimova
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| | | | | | | | | |
Collapse
|
28
|
Abstract
Ouabain, a toxic of vegetal origin used for centuries to treat heart failure, has recently been demonstrated to have an endogenous counterpart, most probably ouabain itself, which behaves as a hormone. Therefore, the challenge now is to discover the physiological role of hormone ouabain. We have recently shown that it modulates cell contacts such as gap junctions, which communicate neighboring cells, as well as tight junctions (TJs), which are one of the two differentiated features of epithelial cells, the other being apical/basolateral polarity. The importance of cell contacts can be hardly overestimated, since the most complex object in the universe, the brain, assembles itself depending on what cells contacts what other(s) how, when, and how is the molecular composition and special arrangement of the contacts involved. In the present chapter, we detail the protocols used to demonstrate the effect of ouabain on the molecular structure and functional properties of one of those cell-cell contacts: the TJ.
Collapse
|
29
|
Rossini GP, Bigiani A. Palytoxin action on the Na(+),K(+)-ATPase and the disruption of ion equilibria in biological systems. Toxicon 2010; 57:429-39. [PMID: 20932855 DOI: 10.1016/j.toxicon.2010.09.011] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 09/03/2010] [Accepted: 09/24/2010] [Indexed: 11/29/2022]
Abstract
Palytoxin-group toxins (PlTX) exert their potent biological activity by altering mechanisms of ion homeostasis in excitable and non-excitable tissues. This review will describe major aspects that led to the relatively early identification of the Na(+),K(+)-ATPase as the molecular target and receptor of the toxin in sensitive systems. The importance of this pump in the normal functioning of animal cells has driven extensive investigative efforts. The recognized molecular mechanism of action of PlTX involves its binding to the extracellular portion of alpha subunit of this plasma membrane protein, which converts an enzyme carrying ions against their concentration gradients at the expense of chemical energy (ATP) into a non-selective cation channel, allowing passive flow of ions following their concentration gradients. More recent findings have indicated that PlTX would interfere with the normal strict coupling between inner and outer gates of the pump controlling the ion access to the Na(+),K(+)-ATPase, allowing the gates to be simultaneously open. The ability of PlTX to make internal portions of the Na(+),K(+)-ATPase accessible to relatively large molecules has been exploited to characterize the structure-function relationship of the pump, leading to a better understanding of its ion translocation pathway. Thus, forty years from the isolation of this potent marine biotoxin, a considerable understanding of its mode of action and of its potential as a research tool have been achieved and are the basis for promising future advancement in the characterization of biological systems and their alteration by PlTX.
Collapse
Affiliation(s)
- Gian Paolo Rossini
- Dipartimento di Scienze Biomediche, Università di Modena e Reggio Emilia, Via G. Campi 287, I-41125 Modena, Italy.
| | | |
Collapse
|
30
|
Abstract
Epithelial cells treated with high concentrations of ouabain (e.g., 1 microM) retrieve molecules involved in cell contacts from the plasma membrane and detach from one another and their substrates. On the basis of this observation, we suggested that ouabain might also modulate cell contacts at low, nontoxic levels (10 or 50 nM). To test this possibility, we analyzed its effect on a particular type of cell-cell contact: the tight junction (TJ). We demonstrate that at concentrations that neither inhibit K(+) pumping nor disturb the K(+) balance of the cell, ouabain modulates the degree of sealing of the TJ as measured by transepithelial electrical resistance (TER) and the flux of neutral 3 kDa dextran (J(DEX)). This modulation is accompanied by changes in the levels and distribution patterns of claudins 1, 2, and 4. Interestingly, changes in TER, J(DEX), and claudins behavior are mediated through signal pathways containing ERK1/2 and c-Src, which have distinct effects on each physiological parameter and claudin type. These observations support the theory that at low concentrations, ouabain acts as a modulator of cell-cell contacts.
Collapse
|
31
|
Padilla-Benavides T, Roldán ML, Larre I, Flores-Benitez D, Villegas-Sepúlveda N, Contreras RG, Cereijido M, Shoshani L. The polarized distribution of Na+,K+-ATPase: role of the interaction between {beta} subunits. Mol Biol Cell 2010; 21:2217-25. [PMID: 20444976 PMCID: PMC2893986 DOI: 10.1091/mbc.e10-01-0081] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Na+,K+-ATPase polarity depends on the interaction between the β subunits of Na+,K+-ATPases located on neighboring cells. In the present work, we use energy transfer methods (FRET), in vivo to demonstrate that these β subunits interact directly at the intercellular space of epithelial cells. The very existence of higher metazoans depends on the vectorial transport of substances across epithelia. A crucial element of this transport is the membrane enzyme Na+,K+-ATPase. Not only is this enzyme distributed in a polarized manner in a restricted domain of the plasma membrane but also it creates the ionic gradients that drive the net movement of glucose, amino acids, and ions across the entire epithelium. In a previous work, we have shown that Na+,K+-ATPase polarity depends on interactions between the β subunits of Na+,K+-ATPases located on neighboring cells and that these interactions anchor the entire enzyme at the borders of the intercellular space. In the present study, we used fluorescence resonance energy transfer and coprecipitation methods to demonstrate that these β subunits have sufficient proximity and affinity to permit a direct interaction, without requiring any additional extracellular molecules to span the distance.
Collapse
Affiliation(s)
- Teresita Padilla-Benavides
- Department of Physiology Biophysics and Neurosciences, and Department of Molecular Biomedicine, Center for Research and Advanced Studies, CINVESTAV-IPN, Mexico DF 07300, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Lingrel JB. The physiological significance of the cardiotonic steroid/ouabain-binding site of the Na,K-ATPase. Annu Rev Physiol 2010; 72:395-412. [PMID: 20148682 DOI: 10.1146/annurev-physiol-021909-135725] [Citation(s) in RCA: 232] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The Na,K-ATPase is the membrane "pump" that generates the Na(+) and K(+) gradients across the plasma membrane that drives many physiological processes. This enzyme is highly sensitive to inhibition by cardiotonic steroids, most notably the digitalis/ouabain class of compounds, which have been used for centuries to treat congestive heart failure and arrhythmias. The amino acids that constitute the ouabain-binding site are highly conserved across the evolutionary spectrum. This could be fortuitous or could result from this site being conserved because it has an important biological function. New physiological approaches using genetically engineered mice are being used to define the biological significance of the "receptor function" of the Na,K-ATPase and its regulation by potential endogenous cardiotonic steroid-like compounds. These studies extend the reach of earlier studies involving the biochemical purification of endogenous regulatory ligands.
Collapse
Affiliation(s)
- Jerry B Lingrel
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0524, USA.
| |
Collapse
|
33
|
Akimova OA, Tremblay J, Van Huysse JW, Hamet P, Orlov SN. Cardiotonic steroid-resistant alpha1-Na+,K+-ATPase rescues renal epithelial cells from the cytotoxic action of ouabain: evidence for a Nai+,Ki+ -independent mechanism. Apoptosis 2010; 15:55-62. [PMID: 19949978 DOI: 10.1007/s10495-009-0429-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mechanisms underlying the tissue-specific impact of cardiotonic steroids (CTS) on cell survival and death remain poorly understood. This study examines the role of Na(+),K(+)-ATPase alpha subunits in death of Madin-Darby canine kidney (MDCK) cells evoked by 24-h exposure to ouabain. MDCK cells expressing a variant of the alpha1 isoform, CTS-sensitive alpha1S, were stably transfected with a cDNA encoding CTS-resistant alpha1R-Na(+),K(+)-ATPase, whose expression was confirmed by RT-PCR. In mock-transfected and alpha1R-cells, maximal inhibition of (86)Rb influx was observed at 10 and 1000 muM ouabain, respectively, thus confirming high abundance of alpha1R-Na(+),K(+)-ATPase in these cells. Six-hour treatment of alpha1R-cells with 1000 muM ouabain led to the same elevation of the [Na(+)](i)/[K(+)](i) ratio that was detected in mock-transfected cells treated with 3 muM ouabain. However, in contrast to the massive death of mock-transfected cells exposed to 3 muM ouabain, alpha1R-cells survived after 24-h incubation with 1000 muM ouabain. Inversion of the [Na(+)](i)/[K(+)](i) ratio evoked by Na(+),K(+)-ATPase inhibition in K(+)-free medium did not affect survival of alpha1R-cells but increased their sensitivity to ouabain. Our results show that the alpha1R subunit rescues MDCK cells from the cytotoxic action of CTS independently of inhibition of Na(+),K(+)-ATPase-mediated Na(+) and K(+) fluxes and inversion of the [Na(+)](i)/[K(+)](i) ratio.
Collapse
Affiliation(s)
- Olga A Akimova
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal, Technopôle Angus, 2901 Rachel Est, Montreal, QC H1W 4A4, Canada.
| | | | | | | | | |
Collapse
|
34
|
Death of ouabain-treated renal epithelial cells: evidence for p38 MAPK-mediated Na (i) (+) /K (i) (+) -independent signaling. Apoptosis 2010; 14:1266-73. [PMID: 19784777 DOI: 10.1007/s10495-009-0404-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent studies demonstrate that cytotoxic actions of ouabain and other cardiotonic steroids (CTS) on renal epithelial cells (REC) are triggered by their interaction with the Na(+),K(+)-ATPase alpha-subunit but not the result of inhibition of Na(+),K(+)-ATPase-mediated ion fluxes and inversion of the [Na(+)](i)/[K(+)](i) ratio. This study examined the role of mitogen-activated protein kinases (MAPK) in the death of ouabain-treated REC. Exposure of C7-MDCK cells that resembled principal cells from canine kidney to 3 microM ouabain led to phosphorylation of p38 without significant impact on phosphorylation of ERK and JNK MAPK. Maximal increment of p38 phosphorylation was observed at 4 h followed by cell death at 12 h of ouabain addition. In contrast to ouabain, neither cell death nor p38 MAPK phosphorylation were affected by elevation of the [Na(+)](i)/[K(+)](i) ratio triggered by Na(+),K(+)-ATPase inhibition in K(+)-free medium. p38 phosphorylation was noted in all other cell types exhibiting death in the presence of ouabain, such as intercalated cells from canine kidney and human colon rectal carcinoma cells. We did not observe any action of ouabain on p38 phosphorylation in ouabain-resistant smooth muscle cells from rat aorta and endothelial cells from human umbilical vein. Both p38 phosphorylation and death of ouabain-treated C7-MDCK cells were suppressed by p38 inhibitor SB 202190 but were resistant to its inactive analogue SB 202474. Our results demonstrate that death of CTS-treated REC is triggered by Na (i) (+) ,K (i) (+) -independent activation of p38 MAPK.
Collapse
|
35
|
Valente RC, Capella LS, Oliveira MMM, Nunes-Lima LT, Cruz FCM, Palmieri RR, Lopes AG, Capella MAM. Diverse actions of ouabain and its aglycone ouabagenin in renal cells. Cell Biol Toxicol 2009; 26:201-13. [PMID: 19757104 DOI: 10.1007/s10565-009-9136-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Accepted: 08/20/2009] [Indexed: 12/15/2022]
Abstract
The cellular actions of ouabain are complex and involve different pathways, depending on the cell type and experimental conditions. Several studies have reported that Madin-Darby canine kidney (MDCK) cellular sensitivity to ouabain is not related to Na-K-ATPase inhibition, and others showed that some cell types, such as Ma104, are resistant to ouabain toxicity albeit their Na-K-ATPase isoforms possess high affinity for this glycoside. We describe here that the effects of ouabain and ouabagenin also diverge in MDCK and Ma104 cells, being MDCK cells more resistant to ouabagenin, while Ma104 cells are resistant to both molecules. This feature seems to correlate with induction of cell signaling, since ouabain, but not ouabagenin, induced an intense and sustained increase in tyrosine phosphorylation levels in MDCK cells. Moreover, ouabain-induced phosphorylation in Ma104 cells was approximately half than that observed in MDCK cells. The proportion between alpha and beta subunits of Na-K-ATPase was similar in MDCK cells, though Ma104 cells presented more alpha subunits, located mainly at the cytoplasm. Furthermore, a fluorescent ouabain-analog labeled mainly the cytoplasm of Ma104 cells, the opposite of that seen in MDCK cells, corroborating the results using anti-Na-K-ATPase antibodies. Hence, the results suggest that ouabain and ouabagenin differ in terms of Na-K-ATPase inhibition and cell signaling activation in MDCK cells. Additionally, MDCK and Ma104 cell lines respond differently to ouabain, perhaps due to an intrinsic ability of this glycoside to selectively reach the cytoplasm of Ma104 cells.
Collapse
Affiliation(s)
- Raphael C Valente
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Fedorova LV, Raju V, El-Okdi N, Shidyak A, Kennedy DJ, Vetteth S, Giovannucci DR, Bagrov AY, Fedorova OV, Shapiro JI, Malhotra D. The cardiotonic steroid hormone marinobufagenin induces renal fibrosis: implication of epithelial-to-mesenchymal transition. Am J Physiol Renal Physiol 2009; 296:F922-34. [PMID: 19176701 DOI: 10.1152/ajprenal.90605.2008] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
We recently demonstrated that the cardiotonic steroid marinobufagenin (MBG) induced fibrosis in rat hearts through direct stimulation of collagen I secretion by cardiac fibroblasts. This stimulation was also responsible for the cardiac fibrosis seen in experimental renal failure. In this study, the effect of MBG on the development of renal fibrosis in rats was investigated. Four weeks of MBG infusion triggered mild periglomerular and peritubular fibrosis in the cortex and the appearance of fibrotic scars in the corticomedullary junction of the kidney. MBG also significantly increased the protein levels and nuclear localization of the transcription factor Snail in the tubular epithelia. It is known that activation of Snail is associated with epithelial-to-mesenchymal transition (EMT) during renal fibrosis. To examine whether MBG alone can trigger EMT, we used the porcine proximal tubular cell line LLC-PK1. MBG (100 nM) caused LLC-PK1 cells grown to confluence to acquire a fibroblast-like shape and have an invasive motility. The expressions of the mesenchymal proteins collagen I, fibronectin, and vimentin were increased twofold. However, the total level of E-cadherin remained unchanged. These alterations in LLC-PK1 cells in the presence of MBG were accompanied by elevated expression and nuclear translocation of Snail. During the time course of EMT, MBG did not have measurable inhibitory effects on the ion pumping activity of its natural ligand, Na(+)-K(+)-ATPase. Our data suggest that the MBG may be an important factor in inducing EMT and, through this mechanism, elevated levels of MBG in chronic renal failure may play a role in the progressive fibrosis.
Collapse
Affiliation(s)
- Larisa V Fedorova
- Division of Nephrology, Dept. of Medicine, Univ. of Toledo College of Medicine, 3000 Arlington Ave., Toledo Ohio, 43614-2598, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Mijatovic T, Ingrassia L, Facchini V, Kiss R. Na+/K+-ATPase alpha subunits as new targets in anticancer therapy. Expert Opin Ther Targets 2009; 12:1403-17. [PMID: 18851696 DOI: 10.1517/14728222.12.11.1403] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND The sodium pump (Na(+)/K(+)-ATPase) could be a target for the development of anticancer drugs as it serves as a signal transducer, it is a player in cell adhesion and its aberrant expression and activity are implicated in the development and progression of different cancers. Cardiotonic steroids (CS) are the natural ligands and inhibitors of the sodium pump and this supports the possibility of their development as anticancer agents targeting overexpressed Na(+)/K(+)-ATPase alpha subunits. OBJECTIVES To highlight and further develop the concept of using Na(+)/K(+)-ATPase alpha1 and alpha3 subunits as targets in anticancer therapy and to address the question of the actual usefulness of further developing CS as anticancer agents. CONCLUSIONS Targeting overexpressed Na(+)/K(+)-ATPase alpha subunits using novel CS might open a new era in anticancer therapy and bring the concept of personalized medicine from aspiration to reality. Clinical data are now needed to further support this proposal. Furthermore, future medicinal chemistry should optimize new anticancer CS to target Na(+)/K(+)-ATPase alpha subunits with the aim of rendering them more potent and less toxic.
Collapse
|
38
|
Louzao MC, Ares IR, Cagide E. Marine toxins and the cytoskeleton: a new view of palytoxin toxicity. FEBS J 2008; 275:6067-74. [PMID: 19016862 DOI: 10.1111/j.1742-4658.2008.06712.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Palytoxin is a marine toxin first isolated from zoanthids (genus Palythoa), even though dinoflagellates of the genus Ostreopsis are the most probable origin of the toxin. Ostreopsis has a wide distribution in tropical and subtropical areas, but recently these dinoflagellates have also started to appear in the Mediterranean Sea. Two of the most remarkable properties of palytoxin are the large and complex structure (with different analogs, such as ostreocin-D or ovatoxin-a) and the extreme acute animal toxicity. The Na(+)/K(+)-ATPase has been proposed as receptor for palytoxin. The marine toxin is known to act on the Na(+) pump and elicit an increase in Na(+) permeability, which leads to depolarization and a secondary Ca(2+) influx, interfering with some functions of cells. Studies on the cellular cytoskeleton have revealed that the signaling cascade triggered by palytoxin leads to actin filament system distortion. The activity of palytoxin on the actin cytoskeleton is only partially associated with the cytosolic Ca(2+) changes; therefore, this ion represents an important factor in altering this structure, but it is not the only cause. The goal of the present minireview is to compile the findings reported to date about: (a) how palytoxin and analogs are able to modify the actin cytoskeleton within different cellular models; and (b) what signaling mechanisms could be involved in the modulation of cytoskeletal dynamics by palytoxin.
Collapse
Affiliation(s)
- M Carmen Louzao
- Departamento de Farmacologia, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo, Spain.
| | | | | |
Collapse
|
39
|
Abstract
The mechanisms of cell death signaling triggered by cardiotonic steroids are poorly understood. Based on massive detachment of ouabain-treated Madin-Darby canine kidney (MDCK) cells, it may be proposed that the cytotoxic action of these compounds is mediated by anoikis, i.e. a particular mode of death occurring in cells lacking cell-to-extracellular matrix interactions. We tested this hypothesis. Six hour incubation of MDCK cells with ouabain, marinobufagenin or K+-free medium almost completely blocked Na+,K+-ATPase, increased Na (i) + content by approximately 10-fold and suppressed cell attachment to regular-plastic-plates by up to 5-fold. In contrast, the death of attached cells was observed after 24-h incubation with ouabain but not in the presence of marinobufagenin or K+-free medium. Cells treated with ouabain and undergoing anoikis on ultra-low attachment plates exhibited different cell volume behaviour, i.e. swelling and shrinkage, respectively. The pan-caspase inhibitor z-VAD.fmk and the protein kinase C activator PMA rescued MDCK cells from anoikis but did not influence the survival of ouabain-treated cells, whereas medium acidification from pH 7.2 to 6.7 almost completely abolished the cytotoxic action of ouabain, but did not significantly affect anoikis. Our results show that the Na (i) + ,K (i) + -independent mode of MDCK cell death evoked by ouabain is not mediated by anoikis.
Collapse
|
40
|
|
41
|
Vagin O, Turdikulova S, Tokhtaeva E. Polarized membrane distribution of potassium-dependent ion pumps in epithelial cells: different roles of the N-glycans of their beta subunits. Cell Biochem Biophys 2007; 47:376-91. [PMID: 17652782 DOI: 10.1007/s12013-007-0033-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 10/23/2022]
Abstract
The Na,K-ATPases and the H,K-ATPases are two potassium-dependent homologous heterodimeric P2-type pumps that catalyze active transport of Na+ in exchange for K+ (Na,K-ATPase) or H+ in exchange for K+ (H,K-ATPase). The ubiquitous Na,K-ATPase maintains intracellular ion balance and membrane potential. The gastric H,K-ATPase is responsible for acid secretion by the parietal cell of the stomach. Both pumps consist of a catalytic alpha-subunit and a glycosylated beta-subunit that is obligatory for normal pump maturation and trafficking. Individual N-glycans linked to the beta-subunits of the Na,K-ATPase and H,K-ATPase are important for stable membrane integration of their respective alpha subunits, folding, stability, subunit assembly, and enzymatic activity of the pumps. They are also essential for the quality control of unassembled beta-subunits that results in either the exit of the subunits from the ER or their ER retention and subsequent degradation. Overall, the importance of N-glycans for the maturation and quality control of the H,K-ATPase is greater than that of the Na,K-ATPase. The roles of individual N-glycans of the beta-subunits in the post-ER trafficking, membrane targeting and plasma membrane retention of the Na,K-ATPase and H,K-ATPase are different. The Na,K-ATPase beta1-subunit is the major beta-subunit isoform in cells with lateral location of the pump. All three N-glycans of the Na,K-ATPase beta1-subunit are important for the lateral membrane retention of the pump due to glycan-mediated interaction between the beta1-subunits of the two neighboring cells in the cell monolayer and cytosolic linkage of the alpha-subunit to the cytoskeleton. This intercellular beta1-beta1 interaction is also important for formation of cell-cell contacts. In contrast, the N-glycans unique to the Na,K-ATPase beta2-subunit,which has up to eight N-glycosylation sites, contain apical sorting information. This is consistent with the apical location of the Na,K-ATPase in normal and malignant epithelial cells with high abundance of the beta2-subunit. Similarly, all seven N-glycans of the gastric H,K-ATPase beta-subunit determine apical sorting of this subunit.
Collapse
Affiliation(s)
- Olga Vagin
- Department of Physiology, School of Medicine, UCLA and Veterans Administration Greater Los Angeles Health Care System, VAGLAHS/West LA, Building 113, Room 324, 11301 Wilshire Blvd, Los Angeles, CA 90073, USA.
| | | | | |
Collapse
|
42
|
Stähli BE, Breitenstein A, Akhmedov A, Camici GG, Shojaati K, Bogdanov N, Steffel J, Ringli D, Lüscher TF, Tanner FC. Cardiac glycosides regulate endothelial tissue factor expression in culture. Arterioscler Thromb Vasc Biol 2007; 27:2769-76. [PMID: 18029910 DOI: 10.1161/atvbaha.107.153502] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Tissue factor (TF) plays an important role in acute coronary syndromes and stent thrombosis. This study investigates whether Na(+)/K(+)-ATPase regulates TF expression in human endothelial cells. METHODS AND RESULTS Ouabain inhibited tumor necrosis factor (TNF)-alpha-induced endothelial TF protein expression; maximal inhibition occurred at 10(-5) mol/L, reached more than 70%, and was observed throughout the 5 hours stimulation period. The decrease in protein expression was paralleled by a reduced TF surface activity. Similarly, lowering of extracellular potassium concentration inhibited TNF-alpha-induced TF protein expression. In contrast, ouabain did not affect TNF-alpha-induced expression of full-length TF mRNA for up to 5 hours of stimulation; instead, expression of alternatively-spliced TF mRNA was upregulated after 3 and 5 hours of stimulation. Ouabain did not affect TNF-alpha-induced activation of the MAP kinases p38, extracellular signal-regulated kinase (ERK), and c-Jun terminal NH(2) kinase; activation of Akt and p70S6 kinase remained unaltered as well. Similar to the MAP kinases, ouabain did not affect TNF-alpha-induced degradation of IkappaB-alpha. Ouabain had no effect on TF protein degradation. CONCLUSIONS Na(+)/K(+)-ATPase is required for protein translation of endothelial TF in culture. This observation provides novel insights into posttranscriptional regulation of TF expression.
Collapse
Affiliation(s)
- Barbara E Stähli
- Cardiovascular Research, Physiology Institute, University of Zürich and Cardiology, University Hospital Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Cereijido M, Contreras RG, Shoshani L, Flores-Benitez D, Larre I. Tight junction and polarity interaction in the transporting epithelial phenotype. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2007; 1778:770-93. [PMID: 18028872 DOI: 10.1016/j.bbamem.2007.09.001] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2007] [Revised: 08/28/2007] [Accepted: 09/03/2007] [Indexed: 12/21/2022]
Abstract
Development of tight junctions and cell polarity in epithelial cells requires a complex cellular machinery to execute an internal program in response to ambient cues. Tight junctions, a product of this machinery, can act as gates of the paracellular pathway, fences that keep the identity of plasma membrane domains, bridges that communicate neighboring cells. The polarization internal program and machinery are conserved in yeast, worms, flies and mammals, and in cell types as different as epithelia, neurons and lymphocytes. Polarization and tight junctions are dynamic features that change during development, in response to physiological and pharmacological challenges and in pathological situations like infection.
Collapse
Affiliation(s)
- Marcelino Cereijido
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV, AP 14-740, México D.F. 07000, México.
| | | | | | | | | |
Collapse
|
44
|
Akimova OA, Hamet P, Orlov SN. [Na+]i/[K+]i -independent death of ouabain-treated renal epithelial cells is not mediated by Na+,K+ -ATPase internalization and de novo gene expression. Pflugers Arch 2007; 455:711-9. [PMID: 17622553 DOI: 10.1007/s00424-007-0283-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2007] [Accepted: 05/02/2007] [Indexed: 10/23/2022]
Abstract
The cytotoxic effect of long-term exposure of renal epithelial cells to ouabain and other cardiotonic steroids (CTS) is mediated by the interaction of these compounds with Na(+),K(+)-ATPase but is independent of the inhibition of Na(+),K(+)-ATPase-mediated ion fluxes. Sustained application of CTS also leads to Na(+),K(+)-ATPase endocytosis and its translocation into the nuclei that might trigger the cell death machinery via the regulation of gene expression. This study examines the role of Na(+),K(+)-ATPase internalization and de novo gene expression in the death of ouabain-treated C7-Madin-Darby canine kidney (MDCK) cells derived from distal tubules of the MDCK. In these cells, 6-h exposure to 3 microM ouabain led to the internalization of approximately 50% of plasmalemmal Na(+),K(+)-ATPase. Prolonged incubation in a K(+)-free medium abolished ouabain-induced Na(+),K(+)-ATPase internalization but did not affect the cytotoxic action of ouabain seen after 18-h incubation. Previously, it was shown that CTS-induced Na(+),K(+)-ATPase internalization is mediated by its interaction with Src within caveolae. Neither caveolae damage by cholesterol depletion with methyl-beta-cyclodextrin nor Src inhibition with 4-amino-5(4-chlorophenyl)-7-(t-butyl)pyrazol[3,4-d]pyridine affected the death of ouabain-treated C7-MDCK cells. Actinomycin D at the 0.1-microg/ml concentration almost completely abolished ribonucleic acid synthesis but did not protect C7-MDCK cells from the cytotoxic action of ouabain. Our results show that neither Na(+),K(+)-ATPase endocytosis nor de novo gene expression contributes to Na(+)(i), K(+)(i)-independent cell death signaling evoked by prolonged exposure to CTS.
Collapse
Affiliation(s)
- Olga A Akimova
- Research Centre, Centre Hospitalier de l'Université de Montréal, Technôpole Angus, 2901 Rachel East, Montreal, Quebec H1W 4A4, Canada
| | | | | |
Collapse
|
45
|
Schoner W, Scheiner-Bobis G. Endogenous and exogenous cardiac glycosides: their roles in hypertension, salt metabolism, and cell growth. Am J Physiol Cell Physiol 2007; 293:C509-36. [PMID: 17494630 DOI: 10.1152/ajpcell.00098.2007] [Citation(s) in RCA: 342] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cardiotonic steroids (CTS), long used to treat heart failure, are endogenously produced in mammals. Among them are the hydrophilic cardenolide ouabain and the more hydrophobic cardenolide digoxin, as well as the bufadienolides marinobufagenin and telecinobufagin. The physiological effects of endogenous ouabain on blood pressure and cardiac activity are consistent with the "Na(+)-lag" hypothesis. This hypothesis assumes that, in cardiac and arterial myocytes, a CTS-induced local increase of Na(+) concentration due to inhibition of Na(+)/K(+)-ATPase leads to an increase of intracellular Ca(2+) concentration ([Ca(2+)](i)) via a backward-running Na(+)/Ca(2+) exchanger. The increase in [Ca(2+)](i) then activates muscle contraction. The Na(+)-lag hypothesis may best explain short-term and inotropic actions of CTS. Yet all data on the CTS-induced alteration of gene expression are consistent with another hypothesis, based on the Na(+)/K(+)-ATPase "signalosome," that describes the interaction of cardiac glycosides with the Na(+) pump as machinery activating various signaling pathways via intramembrane and cytosolic protein-protein interactions. These pathways, which may be activated simultaneously or selectively, elevate [Ca(2+)](i), activate Src and the ERK1/2 kinase pathways, and activate phosphoinositide 3-kinase and protein kinase B (Akt), NF-kappaB, and reactive oxygen species. A recent development indicates that new pharmaceuticals with antihypertensive and anticancer activities may be found among CTS and their derivatives: the antihypertensive rostafuroxin suppresses Na(+) resorption and the Src-epidermal growth factor receptor-ERK pathway in kidney tubule cells. It may be the parent compound of a new principle of antihypertensive therapy. Bufalin and oleandrin or the cardenolide analog UNBS-1450 block tumor cell proliferation and induce apoptosis at low concentrations in tumors with constitutive activation of NF-kappaB.
Collapse
Affiliation(s)
- Wilhelm Schoner
- Institut für Biochemie und Endokrinologie, Fachbereich Veterinärmedizin, Justus-Liebig-Universität Giessen, Frankfurter Str 100, Giessen, Germany.
| | | |
Collapse
|
46
|
Ma L, Kuang K, Smith RW, Rittenband D, Iserovich P, Diecke F, Fischbarg J. Modulation of tight junction properties relevant to fluid transport across rabbit corneal endothelium. Exp Eye Res 2007; 84:790-8. [PMID: 17320078 PMCID: PMC1993899 DOI: 10.1016/j.exer.2006.12.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2006] [Revised: 11/30/2006] [Accepted: 12/15/2006] [Indexed: 11/20/2022]
Abstract
Paracellular junctions could play an important role in corneal endothelial fluid transport. In this study we explored the effects of different reagents on the tight junctional barrier by assessing the translayer specific electrical resistance (TER) across rabbit corneal endothelial preparations and cultured rabbit corneal endothelial cells' (CRCEC) monolayers, the paracellular permeability (Papp) for fluorescein isothiocyanate (FITC) dextrans across CRCEC, and fluid transport across de-epithelialized rabbit corneal endothelial preparations. Palmitoyl carnitine (PC), poly-L-lysine (PLL), adenosine triphosphate (ATP), and dibutyryl adenosine 3',5'-cyclic monophosphate (dB-cAMP) were used to modulate corneal endothelial fluid transport and tight junctions (TJs). After seeding, the TER across CRCEC reached maximal values (29.2+/-1.0 Omega cm2) only after the 10th day. PC (0.1 mM) caused decreases both in TER (by 40%) and fluid transport (swelling rate: 18.5+/-0.3 microm/h), and an increase in Papp. PLL resulted in increased TER rose and Papp but decreased fluid transport (swelling rate: 10+/-0.3 microm/h). dB-cAMP (0.1 mM) and ATP (0.1 mM) decreased TER by 16% and 6%, increased Papp slightly, and stimulated fluid transport; the rates of de-swelling (in microm/h) were -5.4+/-0.3 and -12.1+/-0.4, respectively. PC might cause the junctions to open up unspecifically and thus increase passive leak. PLL is a known junctional charge modifier that may be adding steric hindrance to the tight junctions. The results with dB-cAMP and ATP are consistent with fluid transport via the paracellular route.
Collapse
Affiliation(s)
- Li Ma
- Department of Ophthalmology, College of Physicians and Surgeons, Columbia University
| | - Kunyan Kuang
- Department of Ophthalmology, College of Physicians and Surgeons, Columbia University
| | | | | | - Pavel Iserovich
- Department of Ophthalmology, College of Physicians and Surgeons, Columbia University
| | - F.P.J. Diecke
- Dept. of Pharmacology and Physiology, UMDNJ-New Jersey Medical School, Newark, NJ
| | - Jorge Fischbarg
- Department of Ophthalmology, College of Physicians and Surgeons, Columbia University
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University
| |
Collapse
|
47
|
Nesher M, Shpolansky U, Rosen H, Lichtstein D. The digitalis-like steroid hormones: new mechanisms of action and biological significance. Life Sci 2007; 80:2093-2107. [PMID: 17499813 DOI: 10.1016/j.lfs.2007.03.013] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2007] [Revised: 03/08/2007] [Accepted: 03/14/2007] [Indexed: 12/25/2022]
Abstract
Digitalis-like compounds (DLC) are a family of steroid hormones synthesized in and released from the adrenal gland. DLC, the structure of which resembles that of plant cardiac glycosides, bind to and inhibit the activity of the ubiquitous cell surface enzyme Na(+), K(+)-ATPase. However, there is a large body of evidence suggesting that the regulation of ion transport by Na(+), K(+)-ATPase is not the only physiological role of DLC. The binding of DLC to Na(+), K(+)-ATPase induces the activation of various signal transduction cascades that activate changes in intracellular Ca(++) homeostasis, and in specific gene expression. These, in turn, stimulate endocytosis and affect cell growth and proliferation. At the systemic level, DLC were shown to be involved in the regulation of major physiological parameters including water and salt homeostasis, cardiac contractility and rhythm, systemic blood pressure and behavior. Furthermore, the DLC system has been implicated in several pathological conditions, including cardiac arrhythmias, hypertension, cancer and depressive disorders. This review evaluates the evidence for the different aspects of DLC action and delineates open questions in the field.
Collapse
Affiliation(s)
- Maoz Nesher
- Department of Physiology, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Uri Shpolansky
- Department of Physiology, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Haim Rosen
- The Kuvin Center for the Study of Infectious and Tropical Diseases, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - David Lichtstein
- Department of Physiology, The Hebrew University-Hadassah Medical School, Jerusalem, Israel.
| |
Collapse
|
48
|
Louzao MC, Ares IR, Vieytes MR, Valverde I, Vieites JM, Yasumoto T, Botana LM. The cytoskeleton, a structure that is susceptible to the toxic mechanism activated by palytoxins in human excitable cells. FEBS J 2007; 274:1991-2004. [PMID: 17371505 DOI: 10.1111/j.1742-4658.2007.05743.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Palytoxin is a marine toxin responsible for a fatal type of poisoning in humans named clupeotoxism, with symptoms such as neurologic disturbances. It is believed that it binds to the Na(+)/K(+)-ATPase from the extracellular side and modifies cytosolic ions; nevertheless, its effects on internal cell structures, such as the cytoskeleton, which might be affected by these initial events, have not been fully elucidated. Likewise, ostreocin-D, an analog of palytoxin, has been only recently found, and its action on excitable cells is therefore unknown. Therefore, our aim was to investigate the modifications of ion fluxes associated with palytoxin and ostreocin-D activities, and their effects on an essential cytoskeletal component, the actin system. We used human neuroblastoma cells and fluorescent dyes to detect changes in membrane potential, intracellular Ca(2+) concentration, cell detachment, and actin filaments. Fluorescence values were obtained with spectrofluorymetry, laser-scanning cytometry, and confocal microscopy; the last of these was also used for recording images. Palytoxin and ostreocin-D modified membrane permeability as a first step, triggering depolarization and increasing Ca(2+) influx. The substantial loss of filamentous actin, and the morphologic alterations elicited by both toxins, are possibly secondary to their action on ion channels. The decrease in polymerized actin seemed to be Ca(2+)-independent; however, this ion could be related to actin cytoskeletal organization. Palytoxin and ostreocin-D alter the ion fluxes, targeting pathways that involve the cytoskeletal dynamics of human excitable cells.
Collapse
Affiliation(s)
- M Carmen Louzao
- Departamento de Farmacologia, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo, Spain
| | | | | | | | | | | | | |
Collapse
|
49
|
Khundmiri SJ, Metzler MA, Ameen M, Amin V, Rane MJ, Delamere NA. Ouabain induces cell proliferation through calcium-dependent phosphorylation of Akt (protein kinase B) in opossum kidney proximal tubule cells. Am J Physiol Cell Physiol 2006; 291:C1247-57. [PMID: 16807298 DOI: 10.1152/ajpcell.00593.2005] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiotonic glycosides, like ouabain, inhibit Na+-K+-ATPase. Recent evidence suggests that low molar concentrations of ouabain alter cell growth. Studies were conducted to examine the effect of ouabain on Akt phosphorylation and rate of cell proliferation in opossum kidney (OK) proximal tubule cells. Cells exposed to 10 nM ouabain displayed increased Akt Ser473phosphorylation, as evidenced by an increase in phospho-Akt Ser473band density. Ouabain-stimulated Akt Ser473phosphorylation was inhibited by pretreatment with phosphatidylinositol 3-kinase (PI3K) inhibitors (LY294002 and wortmannin), a PLC inhibitor (edelfosine), and an Akt inhibitor. Moreover, ouabain-mediated Akt Ser473phosphorylation was suppressed by reduction of extracellular calcium (EGTA) or when intracellular calcium was buffered by BAPTA-AM. An inhibitor of calcium store release (TMB-8) and an inhibitor of calcium entry via store-operated calcium channels ( SKF96365 ) also suppressed ouabain-mediated Akt Ser473phosphorylation. In fura-2 AM-loaded cells, 10 nM ouabain increased capacitative calcium entry (CCE). Ouabain at 10 nM did not significantly alter baseline cytoplasmic calcium concentration in control cells. However, treatment with 10 nM ouabain caused a significantly higher ATP-mediated calcium store release. After 24 h, 10 nM ouabain increased the rate of cell proliferation. The Akt inhibitor, BAPTA-AM, SKF96365 , and cyclopiazonic acid suppressed the increase in the rate of cell proliferation caused by 10 nM ouabain. Ouabain at 10 nM caused a detectable increase in86Rb uptake but did not significantly alter Na+-K+-ATPase (ouabain-sensitive pNPPase) activity in crude membranes or cell sodium content. Taken together, the results point to a role for CCE and Akt phosphorylation, in response to low concentrations of ouabain, that increase the rate of cell proliferation without inhibiting Na+-K+-ATPase-mediated ion transport.
Collapse
Affiliation(s)
- Syed J Khundmiri
- Department of Medicine, University of Louisville, Louisville, Kentucky 40202, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Vagin O, Tokhtaeva E, Sachs G. The role of the beta1 subunit of the Na,K-ATPase and its glycosylation in cell-cell adhesion. J Biol Chem 2006; 281:39573-87. [PMID: 17052981 DOI: 10.1074/jbc.m606507200] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Based on recent data showing that overexpression of the Na,K-ATPase beta(1) subunit increased cell-cell adhesion of nonpolarized cells, we hypothesized that the beta(1) subunit can also be involved in the formation of cell-cell contacts in highly polarized epithelial cells. In support of this hypothesis, in Madin-Darby canine kidney (MDCK) cells, the Na,K-ATPase alpha(1) and beta(1) subunits were detected as precisely co-localized with adherens junctions in all stages of the monolayer formation starting from the initiation of cell-cell contact. The Na,K-ATPase and adherens junction protein, beta-catenin, stayed partially co-localized even after their internalization upon disruption of intercellular contacts by Ca(2+) depletion of the medium. The Na,K-ATPase subunits remained co-localized with the adherens junctions after detergent treatment of the cells. In contrast, the heterodimer formed by expressed unglycosylated Na,K-ATPase beta(1) subunit and the endogenous alpha(1) subunit was easily dissociated from the adherens junctions and cytoskeleton by the detergent extraction. The MDCK cell line in which half of the endogenous beta(1) subunits in the lateral membrane were substituted by unglycosylated beta(1) subunits displayed a decreased ability to form cell-to-cell contacts. Incubation of surface-attached MDCK cells with an antibody against the extracellular domain of the Na,K-ATPase beta(1) subunit specifically inhibited cell-cell contact formation. We conclude that the Na,K-ATPase beta(1) subunit is involved in the process of intercellular adhesion and is necessary for association of the heterodimeric Na,K-ATPase with the adherens junctions. Further, normal glycosylation of the Na,K-ATPase beta(1) subunit is essential for the stable association of the pump with the adherens junctions and plays an important role in cell-cell contact formation.
Collapse
Affiliation(s)
- Olga Vagin
- Department of Physiology, School of Medicine, UCLA and Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California 90073, USA
| | | | | |
Collapse
|