1
|
Trant J, Sanchez G, McDermott JP, Blanco G. The cystogenic effects of ouabain in autosomal dominant polycystic kidney disease require cell caveolae. Exp Cell Res 2025; 444:114356. [PMID: 39586486 DOI: 10.1016/j.yexcr.2024.114356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
We have previously shown that the hormone ouabain is a circulating factor which can accelerate the progression of autosomal dominant polycystic kidney disease (ADPKD). At physiologic concentrations, ouabain increases cyst area and fibrosis in kidneys from ADPKD but not wildtype mice. These effects are due to an increased affinity for ouabain by its receptor, Na,K-ATPase (NKA), in the kidneys of ADPKD mice which leads to over-activation of NKA signaling function. Previous studies suggested that ouabain's stimulation of NKA signal transduction is mediated by NKA located within cell caveolae. Here, we determined whether caveolae are involved in the ouabain-induced progression of ADPKD cysts. We generated an ADPKD mouse with a global knockout of the main structural component of caveolae, caveolin-1 (CAV1), which we confirmed lacks caveolae in the kidney. When given physiological amounts of ouabain for 5 months, Pkd1RC/RCCav1-/- mice did not exhibit any changes in cyst progression, contrasting with the Pkd1RC/RC mice which showed a significant increase in cystic area and kidney fibrosis. Also, measures of ouabain-induced cell proliferation, including the number of Ki67-positive nuclei and phosphorylation of the extracellular regulated kinase (ERK) and protein kinase B (Akt), did not increase in the Pkd1RC/RCCav1-/- mice compared with the Pkd1RC/RC mice. Moreover, the abnormally increased affinity for ouabain of NKA in Pkd1RC/RC mice was restored to wildtype levels in the Pkd1RC/RCCav1-/- mice. This work highlights the role of caveolae in ouabain-induced NKA signaling and ADPKD cyst progression.
Collapse
Affiliation(s)
- Jordan Trant
- Department of Cell Biology and Physiology and the Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Gladis Sanchez
- Department of Cell Biology and Physiology and the Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jeffery P McDermott
- Department of Cell Biology and Physiology and the Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Gustavo Blanco
- Department of Cell Biology and Physiology and the Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
2
|
Contreras RG, Torres-Carrillo A, Flores-Maldonado C, Shoshani L, Ponce A. Na +/K +-ATPase: More than an Electrogenic Pump. Int J Mol Sci 2024; 25:6122. [PMID: 38892309 PMCID: PMC11172918 DOI: 10.3390/ijms25116122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
The sodium pump, or Na+/K+-ATPase (NKA), is an essential enzyme found in the plasma membrane of all animal cells. Its primary role is to transport sodium (Na+) and potassium (K+) ions across the cell membrane, using energy from ATP hydrolysis. This transport creates and maintains an electrochemical gradient, which is crucial for various cellular processes, including cell volume regulation, electrical excitability, and secondary active transport. Although the role of NKA as a pump was discovered and demonstrated several decades ago, it remains the subject of intense research. Current studies aim to delve deeper into several aspects of this molecular entity, such as describing its structure and mode of operation in atomic detail, understanding its molecular and functional diversity, and examining the consequences of its malfunction due to structural alterations. Additionally, researchers are investigating the effects of various substances that amplify or decrease its pumping activity. Beyond its role as a pump, growing evidence indicates that in various cell types, NKA also functions as a receptor for cardiac glycosides like ouabain. This receptor activity triggers the activation of various signaling pathways, producing significant morphological and physiological effects. In this report, we present the results of a comprehensive review of the most outstanding studies of the past five years. We highlight the progress made regarding this new concept of NKA and the various cardiac glycosides that influence it. Furthermore, we emphasize NKA's role in epithelial physiology, particularly its function as a receptor for cardiac glycosides that trigger intracellular signals regulating cell-cell contacts, proliferation, differentiation, and adhesion. We also analyze the role of NKA β-subunits as cell adhesion molecules in glia and epithelial cells.
Collapse
Affiliation(s)
| | | | | | | | - Arturo Ponce
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, Mexico City 07360, Mexico; (R.G.C.); (A.T.-C.); (C.F.-M.); (L.S.)
| |
Collapse
|
3
|
Blaustein MP, Hamlyn JM. Sensational site: the sodium pump ouabain-binding site and its ligands. Am J Physiol Cell Physiol 2024; 326:C1120-C1177. [PMID: 38223926 PMCID: PMC11193536 DOI: 10.1152/ajpcell.00273.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/22/2023] [Accepted: 01/10/2024] [Indexed: 01/16/2024]
Abstract
Cardiotonic steroids (CTS), used by certain insects, toads, and rats for protection from predators, became, thanks to Withering's trailblazing 1785 monograph, the mainstay of heart failure (HF) therapy. In the 1950s and 1960s, we learned that the CTS receptor was part of the sodium pump (NKA) and that the Na+/Ca2+ exchanger was critical for the acute cardiotonic effect of digoxin- and ouabain-related CTS. This "settled" view was upended by seven revolutionary observations. First, subnanomolar ouabain sometimes stimulates NKA while higher concentrations are invariably inhibitory. Second, endogenous ouabain (EO) was discovered in the human circulation. Third, in the DIG clinical trial, digoxin only marginally improved outcomes in patients with HF. Fourth, cloning of NKA in 1985 revealed multiple NKA α and β subunit isoforms that, in the rodent, differ in their sensitivities to CTS. Fifth, the NKA is a cation pump and a hormone receptor/signal transducer. EO binding to NKA activates, in a ligand- and cell-specific manner, several protein kinase and Ca2+-dependent signaling cascades that have widespread physiological effects and can contribute to hypertension and HF pathogenesis. Sixth, all CTS are not equivalent, e.g., ouabain induces hypertension in rodents while digoxin is antihypertensinogenic ("biased signaling"). Seventh, most common rodent hypertension models require a highly ouabain-sensitive α2 NKA and the elevated blood pressure is alleviated by EO immunoneutralization. These numerous phenomena are enabled by NKA's intricate structure. We have just begun to understand the endocrine role of the endogenous ligands and the broad impact of the ouabain-binding site on physiology and pathophysiology.
Collapse
Affiliation(s)
- Mordecai P Blaustein
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - John M Hamlyn
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
4
|
Jung H, Zarlenga D, Martin JC, Geldhof P, Hallsworth-Pepin K, Mitreva M. The identification of small molecule inhibitors with anthelmintic activities that target conserved proteins among ruminant gastrointestinal nematodes. mBio 2024; 15:e0009524. [PMID: 38358246 PMCID: PMC10936192 DOI: 10.1128/mbio.00095-24] [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: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Abstract
Gastrointestinal nematode (GIN) infections are a major concern for the ruminant industry worldwide and result in significant production losses. Naturally occurring polyparasitism and increasing drug resistance that potentiate disease outcomes are observed among the most prevalent GINs of veterinary importance. Within the five major taxonomic clades, clade Va represents a group of GINs that predominantly affect the abomasum or small intestine of ruminants. However, the development of effective broad-spectrum anthelmintics against ruminant clade Va GINs has been challenged by a lack of comprehensive druggable genome resources. Here, we first assembled draft genomes for three clade Va species (Cooperia oncophora, Trichostrongylus colubriformis, and Ostertagia ostertagi) and compared them with closely related ruminant GINs. Genome-wide phylogenetic reconstruction showed a relationship among ruminant GINs structured by taxonomic classification. Orthogroup (OG) inference and functional enrichment analyses identified 220 clade Va-specific and Va-conserved OGs, enriched for functions related to cell cycle and cellular senescence. Further transcriptomic analysis identified 61 taxonomically and functionally conserved clade Va OGs that may function as drug targets for new broad-spectrum anthelmintics. Chemogenomic screening identified 11 compounds targeting homologs of these OGs, thus having potential anthelmintic activity. In in vitro phenotypic assays, three kinase inhibitors (digitoxigenin, K-252a, and staurosporine) exhibited broad-spectrum anthelmintic activities against clade Va GINs by obstructing the motility of exsheathed L3 (xL3) or molting of xL3 to L4. These results demonstrate valuable applications of the new ruminant GIN genomes in gaining better insights into their life cycles and offer a contemporary approach to discovering the next generation of anthelmintics.IMPORTANCEGastrointestinal nematode (GIN) infections in ruminants are caused by parasites that inhibit normal function in the digestive tract of cattle, sheep, and goats, thereby causing morbidity and mortality. Coinfection and increasing drug resistance to current therapeutic agents will continue to worsen disease outcomes and impose significant production losses on domestic livestock producers worldwide. In combination with ongoing therapeutic efforts, advancing the discovery of new drugs with novel modes of action is critical for better controlling GIN infections. The significance of this study is in assembling and characterizing new GIN genomes of Cooperia oncophora, Ostertagia ostertagi, and Trichostrongylus colubriformis for facilitating a multi-omics approach to identify novel, biologically conserved drug targets for five major GINs of veterinary importance. With this information, we were then able to demonstrate the potential of commercially available compounds as new anthelmintics.
Collapse
Affiliation(s)
- Hyeim Jung
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dante Zarlenga
- Animal Parasitic Diseases Laboratory, U.S. Department of Agriculture, Agricultural Research Service, Beltsville, Maryland, USA
| | - John C. Martin
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Peter Geldhof
- Laboratory of Parasitology, Faculty of Veterinary Medicine, University of Ghent, Merelbeke, Belgium
| | | | - Makedonka Mitreva
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
5
|
Staehr C, Aalkjaer C, Matchkov V. The vascular Na,K-ATPase: clinical implications in stroke, migraine, and hypertension. Clin Sci (Lond) 2023; 137:1595-1618. [PMID: 37877226 PMCID: PMC10600256 DOI: 10.1042/cs20220796] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 10/26/2023]
Abstract
In the vascular wall, the Na,K-ATPase plays an important role in the control of arterial tone. Through cSrc signaling, it contributes to the modulation of Ca2+ sensitivity in vascular smooth muscle cells. This review focuses on the potential implication of Na,K-ATPase-dependent intracellular signaling pathways in severe vascular disorders; ischemic stroke, familial migraine, and arterial hypertension. We propose similarity in the detrimental Na,K-ATPase-dependent signaling seen in these pathological conditions. The review includes a retrospective proteomics analysis investigating temporal changes after ischemic stroke. The analysis revealed that the expression of Na,K-ATPase α isoforms is down-regulated in the days and weeks following reperfusion, while downstream Na,K-ATPase-dependent cSrc kinase is up-regulated. These results are important since previous studies have linked the Na,K-ATPase-dependent cSrc signaling to futile recanalization and vasospasm after stroke. The review also explores a link between the Na,K-ATPase and migraine with aura, as reduced expression or pharmacological inhibition of the Na,K-ATPase leads to cSrc kinase signaling up-regulation and cerebral hypoperfusion. The review discusses the role of an endogenous cardiotonic steroid-like compound, ouabain, which binds to the Na,K-ATPase and initiates the intracellular cSrc signaling, in the pathophysiology of arterial hypertension. Currently, our understanding of the precise control mechanisms governing the Na,K-ATPase/cSrc kinase regulation in the vascular wall is limited. Understanding the role of vascular Na,K-ATPase signaling is essential for developing targeted treatments for cerebrovascular disorders and hypertension, as the Na,K-ATPase is implicated in the pathogenesis of these conditions and may contribute to their comorbidity.
Collapse
Affiliation(s)
- Christian Staehr
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 35, Aarhus, Denmark
| | - Christian Aalkjaer
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
- Danish Cardiovascular Academy, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
| | - Vladimir V. Matchkov
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
| |
Collapse
|
6
|
Gonçalves-de-Albuquerque CF, Cunha CMCD, Castro LVGD, Martins CDA, Barnese MRC, Burth P, Younes-Ibrahim M. Cellular Pathophysiology of Leptospirosis: Role of Na/K-ATPase. Microorganisms 2023; 11:1695. [PMID: 37512868 PMCID: PMC10383190 DOI: 10.3390/microorganisms11071695] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/04/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
Inada and Ido identified Leptospira sp. as the pathogen responsible for Weil's Disease in 1915. Later, it was confirmed that Leptospira causes leptospirosis. The host microorganism's interaction at the cellular level remained misunderstood for many years. Although different bacterial components have been isolated and purified, the complexity of the molecular interactions between these components and the host and the molecular mechanisms responsible for the systemic dysfunctions still needs to be fully unveiled. Leptospirosis affects virtually all animal species. Its cellular pathophysiology must involve a ubiquitous cellular mechanism in all eukaryotes. Na/K-ATPase is the molecular target of the leptospiral endotoxin (glycolipoprotein-GLP). Na/K-ATPase dysfunctions on different types of cells give rise to the organ disorders manifested in leptospirosis. Concomitantly, the development of a peculiar metabolic disorder characterized by dyslipidemia, with increased levels of circulating free fatty acids and an imbalance in the fatty acid/albumin molar ratio, triggers events of cellular lipotoxicity. Synergistically, multiple molecular stimuli are prompted during the infection, activating inflammasomes and Na/K-ATPase signalosome, leading to pro-inflammatory and metabolic alterations during leptospirosis. Leptospirosis involves diverse molecular mechanisms and alteration in patient inflammatory and metabolic status. Nonetheless, Na/K-ATPase is critical in the disease, and it is targeted by GLP, its components, and other molecules, such as fatty acids, that inhibit or trigger intracellular signaling through this enzyme. Herein, we overview the role of Na/K-ATPase during leptospirosis infection as a potential therapeutic target or an indicator of disease severity.
Collapse
Affiliation(s)
| | - Carolina Medina Coeli da Cunha
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro (UNIRIO), Rio de Janeiro 20211-030, Brazil
- Neuroscience Graduate Program, Federal Fluminense University (UFF), Niteroi 24000-000, Brazil
| | | | - Caroline de Azevedo Martins
- School of Medicine and Surgery, Federal University of the State of Rio de Janeiro (UNIRIO), Rio de Janeiro 20270-901, Brazil
| | | | - Patrícia Burth
- Laboratory of Enzymology and Cellular Signaling, Department of Cellular and Molecular Biology, Federal Fluminense University (UFF), Niteroi 24000-000, Brazil
| | - Mauricio Younes-Ibrahim
- FISCLINEX Postgraduate Program, State University of Rio de Janeiro (UERJ), Rio de Janeiro 20550-900, Brazil
- Department of Medicine, Pontifical Catholic University of Rio de Janeiro (PUC-Rio), Rio de Janeiro 22453-900, Brazil
- State University of Rio de Janeiro (UERJ), Rio de Janeiro 20550-900, Brazil
| |
Collapse
|
7
|
Yang K, Li Z, Chen Y, Yin F, Ji X, Zhou J, Li X, Zeng T, Fei C, Ren C, Wang Y, Fang L, Chen L, Zhang P, Mu L, Qian Y, Chen Y, Yin W. Na, K-ATPase α1 cooperates with its endogenous ligand to reprogram immune microenvironment of lung carcinoma and promotes immune escape. SCIENCE ADVANCES 2023; 9:eade5393. [PMID: 36763655 PMCID: PMC9916986 DOI: 10.1126/sciadv.ade5393] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/11/2023] [Indexed: 06/18/2023]
Abstract
Dysregulated endocrine hormones (EHs) contribute to tumorigenesis, but how EHs affect the tumor immune microenvironment (TIM) and the immunotherapy of non-small cell lung cancer (NSCLC) is still unclear. Here, endogenous ouabain (EO), an adrenergic hormone, is elevated in patients with NSCLC and closely related to tumor pathological stage, metastasis, and survival. EO promotes the suppression of TIM in vivo by modulating the expression of immune checkpoint proteins, in which programmed cell death protein ligand 1 (PD-L1) plays a major role. EO increases PD-L1 transcription; however, the EO receptor Na- and K-dependent adenosine triphosphatase (Na, K-ATPase) α1 interacts with PD-L1 to trigger the endocytic degradation of PD-L1. This seemingly contradictory result led us to discover the mechanism whereby EO cooperates with Na, K-ATPase α1 to finely control PD-L1 expression and dampen tumoral immunity. In conclusion, the Na, K-ATPase α1/EO signaling facilitates immune escape in lung cancer, and manipulation of this signaling shows great promise in improving immunotherapy for lung adenocarcinoma.
Collapse
Affiliation(s)
- Kaiyong Yang
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zijian Li
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Yan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Fangzhou Yin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaojun Ji
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jiaqian Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Xin Li
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Tao Zeng
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Chenghao Fei
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chenchen Ren
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yulin Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lei Fang
- Jiangsu Key Laboratory of Molecular Medicine, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Lili Chen
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Pei Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Liyan Mu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuxuan Qian
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Yan Chen
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Wu Yin
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| |
Collapse
|
8
|
Rognant S, Kravtsova VV, Bouzinova EV, Melnikova EV, Krivoi II, Pierre SV, Aalkjaer C, Jepps TA, Matchkov VV. The microtubule network enables Src kinase interaction with the Na,K-ATPase to generate Ca2+ flashes in smooth muscle cells. Front Physiol 2022; 13:1007340. [PMID: 36213229 PMCID: PMC9538378 DOI: 10.3389/fphys.2022.1007340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 09/05/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Several local Ca2+ events are characterized in smooth muscle cells. We have previously shown that an inhibitor of the Na,K-ATPase, ouabain induces spatially restricted intracellular Ca2+ transients near the plasma membrane, and suggested the importance of this signaling for regulation of intercellular coupling and smooth muscle cell contraction. The mechanism behind these Na,K-ATPase-dependent “Ca2+ flashes” remains to be elucidated. In addition to its conventional ion transport function, the Na,K-ATPase is proposed to contribute to intracellular pathways, including Src kinase activation. The microtubule network is important for intracellular signaling, but its role in the Na,K-ATPase-Src kinase interaction is not known. We hypothesized the microtubule network was responsible for maintaining the Na,K-ATPase-Src kinase interaction, which enables Ca2+ flashes. Methods: We characterized Ca2+ flashes in cultured smooth muscle cells, A7r5, and freshly isolated smooth muscle cells from rat mesenteric artery. Cells were loaded with Ca2+-sensitive fluorescent dyes, Calcium Green-1/AM and Fura Red/AM, for ratiometric measurements of intracellular Ca2+. The Na,K-ATPase α2 isoform was knocked down with siRNA and the microtubule network was disrupted with nocodazole. An involvement of the Src signaling was tested pharmacologically and with Western blot. Protein interactions were validated with proximity ligation assays. Results: The Ca2+ flashes were induced by micromolar concentrations of ouabain. Knockdown of the α2 isoform Na,K-ATPase abolished Ca2+ flashes, as did inhibition of tyrosine phosphorylation with genistein and PP2, and the inhibitor of the Na,K-ATPase-dependent Src activation, pNaKtide. Ouabain-induced Ca2+ flashes were associated with Src kinase activation by phosphorylation. The α2 isoform Na,K-ATPase and Src kinase colocalized in the cells. Disruption of microtubule with nocodazole inhibited Ca2+ flashes, reduced Na,K-ATPase/Src interaction and Src activation. Conclusion: We demonstrate that the Na,K-ATPase-dependent Ca2+ flashes in smooth muscle cells require an interaction between the α2 isoform Na, K-ATPase and Src kinase, which is maintained by the microtubule network.
Collapse
Affiliation(s)
- Salomé Rognant
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Violetta V. Kravtsova
- Department of General Physiology, St. Petersburg State University, St. Petersburg, Russia
| | | | | | - Igor I. Krivoi
- Department of General Physiology, St. Petersburg State University, St. Petersburg, Russia
| | - Sandrine V. Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV, United States
| | | | - Thomas A. Jepps
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vladimir V. Matchkov
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- *Correspondence: Vladimir V. Matchkov,
| |
Collapse
|
9
|
Liu J, Chaudhry M, Bai F, Chuang J, Chaudhry H, Al-Astal AEY, Nie Y, Sollars V, Sodhi K, Seligman P, Shapiro JI. Blockage of the Na-K-ATPase signaling-mediated oxidant amplification loop elongates red blood cell half-life and ameliorates uremic anemia induced by 5/6th PNx in C57BL/6 mice. Am J Physiol Renal Physiol 2022; 322:F655-F666. [PMID: 35435001 PMCID: PMC9076417 DOI: 10.1152/ajprenal.00189.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 03/21/2022] [Accepted: 04/06/2022] [Indexed: 11/22/2022] Open
Abstract
We have previously demonstrated that the Na-K-ATPase signaling-mediated oxidant amplification loop contributes to experimental uremic cardiomyopathy and anemia induced by 5/6th partial nephrectomy (PNx). This process can be ameliorated by systemic administration of the peptide pNaKtide, which was designed to block this oxidant amplification loop. The present study demonstrated that the PNx-induced anemia is characterized by marked decreases in red blood cell (RBC) survival as assessed by biotinylated RBC clearance and eryptosis as assessed by annexin V binding. No significant change in iron homeostasis was observed. Examination of plasma samples demonstrated that PNx induced significant increases in systemic oxidant stress as assessed by protein carbonylation, plasma erythropoietin concentration, and blood urea nitrogen. Systemic administration of pNaKtide, but not NaKtide (pNaKtide without the TAT leader sequence) and a scramble "pNaKtide" (sc-pNaKtide), led to the normalization of hematocrit, RBC survival, and plasma protein carbonylation. Administration of the three peptides had no significant effect on PNx-induced increases in plasma erythropoietin and blood urea nitrogen without notable changes in iron metabolism. These data indicate that blockage of the Na-K-ATPase signaling-mediated oxidant amplification loop ameliorates the anemia of experimental renal failure by increasing RBC survival.NEW & NOTEWORTHY The anemia of CKD is multifactorial, and the current treatment based primarily on stimulating bone marrow production of RBCs with erythropoietin or erythropoietin analogs is unsatisfactory. In a murine model of CKD that is complicated by anemia, blockade of Na-K-ATPase signaling with a specific peptide (pNaKtide) ameliorated the anemia primarily by increasing RBC survival. Should these results be confirmed in patients, this strategy may allow for novel and potentially additive strategies to treat the anemia of CKD.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Muhammad Chaudhry
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Fang Bai
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Justin Chuang
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Hibba Chaudhry
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Ala-Eddin Yassin Al-Astal
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Ying Nie
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Vincent Sollars
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Komal Sodhi
- Department of Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Paul Seligman
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Joseph I Shapiro
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| |
Collapse
|
10
|
Shen Y, Cai H, Ma S, Zhu W, Zhao H, Li J, Ye H, Yang L, Zhao C, Huang X, Xiao Z. Telocinobufagin Has Antitumor Effects in Non-Small-Cell Lung Cancer by Inhibiting STAT3 Signaling. JOURNAL OF NATURAL PRODUCTS 2022; 85:765-775. [PMID: 35200033 DOI: 10.1021/acs.jnatprod.1c00761] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Non-small-cell lung carcer (NSCLC), the main histological subtype of lung cancer, is responsible for significant morbidity and mortality worldwide. Telocinobufagin, an active compound of the Chinese traditional medicine ChanSu, has antitumor effects, but its mechanism of action remains unknown. Therefore, we investigated the effect of telocinobufagin on NSCLC growth and metastasis and its possible mechanism of action, in vitro and in vivo. Cell proliferation, migration, and apoptosis were measured by methyl thiazol tetrazolium assay, colony formation, 5-ethynyl-2'-deoxyuridine incorporation, Transwell migration, wound healing, and flow cytometry analysis. A mouse xenograft model was used to evaluate tumor formation in vivo. Telocinobufagin was found to suppress proliferation and metastasis and induce apoptosis in human NSCLC cells. Moreover, telocinobufagin was able to significantly inhibit STAT3 phosphorylation at tyrosine 705 (Y705) and its downstream targets. Additionally, telocinobufagin also impaired the IL-6-induced nuclear translocation of STAT3. Consistent with the in vitro experiments, telocinobufagin reduced the A549 xenograft tumor burden and the levels of P-STAT3Y705, MCL1, BCL2, and cleaved PARP1 in vivo. These results support telocinobufagin as a promising STAT3 signaling inhibitor candidate for the treatment of NSCLC patients.
Collapse
Affiliation(s)
- Yili Shen
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, China
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Haijian Cai
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, China
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Shenjie Ma
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, China
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Wenjing Zhu
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, China
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Haiyang Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jifa Li
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, China
| | - Hua Ye
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, China
| | - Lehe Yang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Chengguang Zhao
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, China
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaoying Huang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Zhongxiao Xiao
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, China
| |
Collapse
|
11
|
Liu J, Tian J, Sodhi K, Shapiro JI. The Na/K-ATPase Signaling and SGLT2 Inhibitor-Mediated Cardiorenal Protection: A Crossed Road? J Membr Biol 2021; 254:513-529. [PMID: 34297135 PMCID: PMC8595165 DOI: 10.1007/s00232-021-00192-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/06/2021] [Indexed: 12/17/2022]
Abstract
In different large-scale clinic outcome trials, sodium (Na+)/glucose co-transporter 2 (SGLT2) inhibitors showed profound cardiac- and renal-protective effects, making them revolutionary treatments for heart failure and kidney disease. Different theories are proposed according to the emerging protective effects other than the original purpose of glucose-lowering in diabetic patients. As the ATP-dependent primary ion transporter providing the Na+ gradient to drive other Na+-dependent transporters, the possible role of the sodium–potassium adenosine triphosphatase (Na/K-ATPase) as the primary ion transporter and its signaling function is not explored.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Biomedical Sciences, JCE School of Medicine, Marshall University, Huntington, WV, USA.
| | - Jiang Tian
- Department of Biomedical Sciences, JCE School of Medicine, Marshall University, Huntington, WV, USA
| | - Komal Sodhi
- Department of Surgery, JCE School of Medicine, Marshall University, Huntington, WV, USA
| | - Joseph I Shapiro
- Departments of Medicine, JCE School of Medicine, Marshall University, Huntington, WV, USA
| |
Collapse
|
12
|
Wang J, Wang X, Gao Y, Lin Z, Chen J, Gigantelli J, Shapiro JI, Xie Z, Pierre SV. Stress Signal Regulation by Na/K-ATPase As a New Approach to Promote Physiological Revascularization in a Mouse Model of Ischemic Retinopathy. Invest Ophthalmol Vis Sci 2021; 61:9. [PMID: 33275652 PMCID: PMC7718810 DOI: 10.1167/iovs.61.14.9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose The identification of target pathways to block excessive angiogenesis while simultaneously restoring physiological vasculature is an unmet goal in the therapeutic management of ischemic retinopathies. pNaKtide, a cell-permeable peptide that we have designed by mapping the site of α1 Na/K-ATPase (NKA)/Src binding, blocks the formation of α1 NKA/Src/reactive oxygen species (ROS) amplification loops and restores physiological ROS signaling in a number of oxidative disease models. The aim of this study was to evaluate the importance of the NKA/Src/ROS amplification loop and the effect of pNaKtide in experimental ischemic retinopathy. Methods Human retinal microvascular endothelial cells (HRMECs) and retinal pigment epithelium (ARPE-19) cells were used to evaluate the effect of pNaKtide on viability, proliferation, and angiogenesis. Retinal toxicity and distribution were assessed in those cells and in the mouse. Subsequently, the role and molecular mechanism of NKA/Src in ROS stress signaling were evaluated biochemically in the retinas of mice exposed to the well-established protocol of oxygen-induced retinopathy (OIR). Finally, pNaKtide efficacy was assessed in this model. Results The results suggest a key role of α1 NKA in the regulation of ROS stress and the Nrf2 pathway in mouse OIR retinas. Inhibition of α1 NKA/Src by pNaKtide reduced pathologic ROS signaling and restored normal expression of hypoxia-inducible factor 1-α/vascular endothelial growth factor (VEGF). Unlike anti-VEGF agents, pNaKtide did promote retinal revascularization while inhibiting neovascularization and inflammation. Conclusions Targeting α1 NKA represents a novel strategy to develop therapeutics that not only inhibit neovascularization but also promote physiological revascularization in ischemic eye diseases.
Collapse
Affiliation(s)
- Jiayan Wang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States.,Departments of Medicine, Ophthalmology, Pharmacology, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Xiaoliang Wang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States.,Departments of Medicine, Ophthalmology, Pharmacology, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Yingnyu Gao
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Zhucheng Lin
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Jing Chen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - James Gigantelli
- Departments of Medicine, Ophthalmology, Pharmacology, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Joseph I Shapiro
- Departments of Medicine, Ophthalmology, Pharmacology, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| |
Collapse
|
13
|
Chen L, Tang S, Li X, Kuang Y, Huang H, Fan P, Feng F, Liu W. A review on traditional usages, chemical constituents and pharmacological activities of periploca forrestii schltr. JOURNAL OF ETHNOPHARMACOLOGY 2021; 271:113892. [PMID: 33516929 DOI: 10.1016/j.jep.2021.113892] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 01/13/2021] [Accepted: 01/26/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Periploca forrestii Schltr. was listed as a classical medicinal plant in "Miao medicine", which is a branch of traditional Chinese medicine (TCM). According to the theory of TCM, P. forrestii has the efficacy of relaxing tendons and activating collaterals, and dispelling wind and eliminating dampness. Hence, it was often used for the therapy of rheumatoid arthritis and traumatic injury in clinical practice. AIMS OF THE REVIEW This review aims to present comprehensive information for the research progress of P. forrestii. The researches on botany, traditional uses, phytochemistry, pharmacology and toxicology of the plant are summarized. We mainly focus on the phytochemical and pharmacological investigations. As a representative class of phytochemicals in P. forrestii, more attention is paid to cardiac glycosides. The insights into potential action of mechanisms and possible future studies on P. forrestii are also discussed. MATERIALS AND METHODS Relevant literature was acquired from scientific databases including Google Scholar, Web of Science, Scifinder, Baidu Scholar, PubMed and Chinese national knowledge infrastructure. Monographs and Chinese pharmacopoeia were also utilized as references. RESULTS To date, all kinds of phytochemical constituents have been isolated and identified from this plant including cardiac glycosides, steroids, terpenoids, flavonoids, phenylpropanoids, quinones, organic phenolic acids and others. Among these, cardiac glycosides were considered as the major ingredients and bioactive materials. Modern pharmacological studies demonstrated that the plant possessed extensive bioactivity, such as anti-inflammatory and analgesic effects, immunosuppressive action, wound healing activity, antioxidant, anti-tumor and, cardiotonic properties. CONCLUSIONS As an important medicinal plant, lots of studies have proved that P. forrestii has significant therapeutical effects, especially on rheumatoid arthritis and traumatic injury. These results provide modern scientific evidence for traditional use and contribute to the development of novel remedies for chronic diseases. However, the exact mechanism of action remains to be elucidated. Furthermore, the long-term in vivo toxicity and clinical efficacy also require in-depth exploration in the future.
Collapse
Affiliation(s)
- Lei Chen
- National Engineering Research Center for Modernization of Traditional Chinese Medicine- Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University,Ganzhou, 341000, China
| | - Siqi Tang
- National Engineering Research Center for Modernization of Traditional Chinese Medicine- Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University,Ganzhou, 341000, China
| | - Xiaojun Li
- National Engineering Research Center for Modernization of Traditional Chinese Medicine- Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University,Ganzhou, 341000, China
| | - Ying Kuang
- National Engineering Research Center for Modernization of Traditional Chinese Medicine- Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University,Ganzhou, 341000, China
| | - Hao Huang
- National Engineering Research Center for Modernization of Traditional Chinese Medicine- Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University,Ganzhou, 341000, China
| | - Pinglong Fan
- National Engineering Research Center for Modernization of Traditional Chinese Medicine- Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University,Ganzhou, 341000, China
| | - Feng Feng
- Jiangsu Food and Pharmaceutical Science College, Huaian, 223003, China.
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
14
|
Souza E Souza KFC, Moraes BPT, Paixão ICNDP, Burth P, Silva AR, Gonçalves-de-Albuquerque CF. Na +/K +-ATPase as a Target of Cardiac Glycosides for the Treatment of SARS-CoV-2 Infection. Front Pharmacol 2021; 12:624704. [PMID: 33935717 PMCID: PMC8085498 DOI: 10.3389/fphar.2021.624704] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/22/2021] [Indexed: 12/11/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), identified for the first time in Wuhan, China, causes coronavirus disease 2019 (COVID-19), which moved from epidemic status to becoming a pandemic. Since its discovery in December 2019, there have been countless cases of mortality and morbidity due to this virus. Several compounds such as chloroquine, hydroxychloroquine, lopinavir-ritonavir, and remdesivir have been tested as potential therapies; however, no effective treatment is currently recommended by regulatory agencies. Some studies on respiratory non-enveloped viruses such as adenoviruses and rhinovirus and some respiratory enveloped viruses including human respiratory syncytial viruses, influenza A, parainfluenza, SARS-CoV, and SARS-CoV-2 have shown the antiviral activity of cardiac glycosides, correlating their effect with Na+/K+-ATPase (NKA) modulation. Cardiac glycosides are secondary metabolites used to treat patients with cardiac insufficiency because they are the most potent inotropic agents. The effects of cardiac glycosides on NKA are dependent on cell type, exposure time, and drug concentration. They may also cause blockage of Na+ and K+ ionic transport or trigger signaling pathways. The antiviral activity of cardiac glycosides is related to cell signaling activation through NKA inhibition. Nuclear factor kappa B (NFκB) seems to be an essential transcription factor for SARS-CoV-2 infection. NFκB inhibition by cardiac glycosides interferes directly with SARS-CoV-2 yield and inflammatory cytokine production. Interestingly, the antiviral effect of cardiac glycosides is associated with tyrosine kinase (Src) activation, and NFκB appears to be regulated by Src. Src is one of the main signaling targets of the NKA α-subunit, modulating other signaling factors that may also impair viral infection. These data suggest that Src-NFκB signaling modulated by NKA plays a crucial role in the inhibition of SARS-CoV-2 infection. Herein, we discuss the antiviral effects of cardiac glycosides on different respiratory viruses, SARS-CoV-2 pathology, cell signaling pathways, and NKA as a possible molecular target for the treatment of COVID-19.
Collapse
Affiliation(s)
- Kauê Francisco Corrêa Souza E Souza
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil.,Laboratório de Imunofarmacologia, Departamento de Bioquímica, Universidade Federal Do Estado Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bianca Portugal Tavares Moraes
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil.,Programa de Pós-Graduação Em Neurociências (PPGNEURO), Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Izabel Christina Nunes de Palmer Paixão
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil.,Programa de Pós-Graduação Em Ciências e Biotecnologia (PPBI), Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil.,Programa de Pós-Graduação Em Neûrologia/Neurociências, Hospital Antônio Pedro Universidade Federal Fluminense, Niterói, Brazil
| | - Patrícia Burth
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil.,Programa de Pós-Graduação Em Ciências e Biotecnologia (PPBI), Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Adriana Ribeiro Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil.,Programa de Pós-Graduação Em Neurociências (PPGNEURO), Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratório de Imunofarmacologia, Departamento de Bioquímica, Universidade Federal Do Estado Do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil.,Programa de Pós-Graduação Em Neurociências (PPGNEURO), Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil.,Programa de Pós-Graduação Em Ciências e Biotecnologia (PPBI), Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil.,Programa de Pós-Graduação Em Biologia Celular e Molecular (PPGBMC), Universidade Federal Do Estado Do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
15
|
Parreira GM, Faria JA, Marques SMS, Garcia IJP, Silva IF, De Carvalho LED, Villar JAFP, Machado MV, de Castro Lima M, Barbosa LA, Cortes VF, de Lima Santos H. The γ-Benzylidene Digoxin Derivative BD-15 Increases the α3-Na, K-ATPase Activity in Rat Hippocampus and Prefrontal Cortex and no Change on Heart. J Membr Biol 2021; 254:189-199. [PMID: 33598793 DOI: 10.1007/s00232-021-00173-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023]
Abstract
Our study aimed to investigate the effects of the new cardiotonic steroid BD-15 (γ-benzylidene derivatives) in the behavioral parameters, oxidative stress and the Na, K-ATPase activity in the hippocampus, prefrontal cortex and heart from rats to verify the safety and possible utilization in brain disorders. For this study, groups of male Wistar rats were used after intraperitoneal injection of 20, 100 and 200 µg/Kg with BD-15. The groups were treated for three consecutive days and the control group received 0.9% saline. BD-15 did not alter behavior of rats treated with different doses. An increase in the specific α2,3-Na, K-ATPase activity was observed for all doses of BD-15 tested in the hippocampus. However, in the prefrontal cortex, only the dose of 100 µg/Kg increased the activity of all Na, K-ATPase isoforms. BD-15 did not cause alteration in the lipid peroxidation levels in the hippocampus, but in the prefrontal cortex, a decrease of lipid peroxidation (~ 25%) was observed. In the hippocampus, GSH levels increased with all doses tested, while in the prefrontal cortex no changes were found. Subsequently, when the effect of BD-15 on cardiac tissue was analyzed, no changes were observed in the tested parameters. BD-15 at a dosage of 100 µg/Kg proved to be promising because it is considered therapeutic for brain disorders, since it increases the activity of the α3-Na, K-ATPase in the hippocampus and prefrontal cortex, as well as decreasing the oxidative stress in these brain regions. In addition, this drug did not cause changes in the tissues of the heart and kidneys, preferentially demonstrating specificity for the brain.
Collapse
Affiliation(s)
- Gabriela Machado Parreira
- Laboratório de Bioquímica Celular, Universidade Federal de São João Del Rei, Campus Centro-Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Divinopolis, MG, Zip Code: 35501-296, Brazil
| | - Jéssica Alves Faria
- Laboratório de Anatomia Humana, Universidade Federal de São João Del Rei, Campus Centro-Oeste Dona Lindu, Divinopolis, MG, Brazil
| | - Sarah Melo Silva Marques
- Laboratório de Bioquímica Celular, Universidade Federal de São João Del Rei, Campus Centro-Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Divinopolis, MG, Zip Code: 35501-296, Brazil
| | - Israel José Pereira Garcia
- Laboratório de Bioquímica Celular, Universidade Federal de São João Del Rei, Campus Centro-Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Divinopolis, MG, Zip Code: 35501-296, Brazil
| | - Isabella Ferreira Silva
- Laboratório de Processamento de Tecidos, Universidade Federal de São João Del Rei, Campus Centro-Oeste Dona Lindu, Divinopolis, MG, Brazil
| | - Luciana Estefani Drumond De Carvalho
- Laboratório de Bioquímica Celular, Universidade Federal de São João Del Rei, Campus Centro-Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Divinopolis, MG, Zip Code: 35501-296, Brazil
| | - José Augusto Ferreira Perez Villar
- Laboratório de Síntese Orgânica e Nanoestruturas, Universidade Federal de São João Del Rei, Campus Centro-Oeste Dona Lindu, Divinopolis, MG, Brazil
| | - Matthews Vieira Machado
- Laboratório de Síntese Orgânica e Nanoestruturas, Universidade Federal de São João Del Rei, Campus Centro-Oeste Dona Lindu, Divinopolis, MG, Brazil
| | - Maira de Castro Lima
- Laboratório de Anatomia Humana, Universidade Federal de São João Del Rei, Campus Centro-Oeste Dona Lindu, Divinopolis, MG, Brazil
| | - Leandro Augusto Barbosa
- Laboratório de Bioquímica Celular, Universidade Federal de São João Del Rei, Campus Centro-Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Divinopolis, MG, Zip Code: 35501-296, Brazil
| | - Vanessa Faria Cortes
- Laboratório de Bioquímica Celular, Universidade Federal de São João Del Rei, Campus Centro-Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Divinopolis, MG, Zip Code: 35501-296, Brazil
| | - Hérica de Lima Santos
- Laboratório de Bioquímica Celular, Universidade Federal de São João Del Rei, Campus Centro-Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Divinopolis, MG, Zip Code: 35501-296, Brazil.
| |
Collapse
|
16
|
Xu Y, Marck P, Huang M, Xie JX, Wang T, Shapiro JI, Cai L, Feng F, Xie Z. Biased Effect of Cardiotonic Steroids on Na/K-ATPase-Mediated Signal Transduction. Mol Pharmacol 2021; 99:217-225. [PMID: 33495275 DOI: 10.1124/molpharm.120.000101] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 12/24/2020] [Indexed: 01/13/2023] Open
Abstract
Recent studies have revealed that Na/K-ATPase (NKA) can transmit signals through ion-pumping-independent activation of pathways relayed by distinct intracellular protein/lipid kinases, and endocytosis challenges the traditional definition that cardiotonic steroids (CTS) are NKA inhibitors. Although additional effects of CTS have long been suspected, revealing its agonist impact through the NKA receptor could be a novel mechanism in understanding the basic biology of NKA. In this study, we tested whether different structural CTS could trigger different sets of NKA/effector interactions, resulting in biased signaling responses without compromising ion-pumping capacity. Using purified NKA, we found that ouabain, digitoxigenin, and somalin cause comparable levels of NKA inhibition. However, although endogenous ouabain stimulates both protein kinases and NKA endocytosis, digitoxigenin and somalin bias to protein kinases and endocytosis, respectively, in LLC-PK1 cells. The positive inotropic effects of CTS are traditionally regarded as NKA inhibitors. However, CTS-induced signaling occurs at concentrations at least one order of magnitude lower than that of inotropy, which eliminates their well known toxic actions on the heart. The current study adds a novel mechanism that CTS could exert its biased signaling properties through the NKA signal transducer. SIGNIFICANCE STATEMENT: Although it is now well accepted that NKA has an ion-pumping-independent signaling function, it is still debated whether direct and conformation-dependent NKA/effector interaction is a key to this function. Therefore, this investigation is significant in advancing our understanding of the basic biology of NKA-mediated signal transduction and gaining molecular insight into the structural elements that are important for cardiotonic steroid's biased action.
Collapse
Affiliation(s)
- Yunhui Xu
- Marshall Institute for Interdisciplinary Research, Huntington, West Virginia (Y.X., P.M., M.H., T.W., L.C., Z.X.); University of Toledo College of Medicine and Life Sciences, Toledo, Ohio (J.X.X.); Joan C. Edwards School of Medicine at Marshall University, Huntington, West Virginia (J.I.S.); and Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, P. R. China, and Jiangsu Food and Pharmaceutical Science College, Huai'an, P. R. China (F.F.)
| | - Pauline Marck
- Marshall Institute for Interdisciplinary Research, Huntington, West Virginia (Y.X., P.M., M.H., T.W., L.C., Z.X.); University of Toledo College of Medicine and Life Sciences, Toledo, Ohio (J.X.X.); Joan C. Edwards School of Medicine at Marshall University, Huntington, West Virginia (J.I.S.); and Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, P. R. China, and Jiangsu Food and Pharmaceutical Science College, Huai'an, P. R. China (F.F.)
| | - Minqi Huang
- Marshall Institute for Interdisciplinary Research, Huntington, West Virginia (Y.X., P.M., M.H., T.W., L.C., Z.X.); University of Toledo College of Medicine and Life Sciences, Toledo, Ohio (J.X.X.); Joan C. Edwards School of Medicine at Marshall University, Huntington, West Virginia (J.I.S.); and Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, P. R. China, and Jiangsu Food and Pharmaceutical Science College, Huai'an, P. R. China (F.F.)
| | - Jeffrey X Xie
- Marshall Institute for Interdisciplinary Research, Huntington, West Virginia (Y.X., P.M., M.H., T.W., L.C., Z.X.); University of Toledo College of Medicine and Life Sciences, Toledo, Ohio (J.X.X.); Joan C. Edwards School of Medicine at Marshall University, Huntington, West Virginia (J.I.S.); and Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, P. R. China, and Jiangsu Food and Pharmaceutical Science College, Huai'an, P. R. China (F.F.)
| | - Tong Wang
- Marshall Institute for Interdisciplinary Research, Huntington, West Virginia (Y.X., P.M., M.H., T.W., L.C., Z.X.); University of Toledo College of Medicine and Life Sciences, Toledo, Ohio (J.X.X.); Joan C. Edwards School of Medicine at Marshall University, Huntington, West Virginia (J.I.S.); and Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, P. R. China, and Jiangsu Food and Pharmaceutical Science College, Huai'an, P. R. China (F.F.)
| | - Joseph I Shapiro
- Marshall Institute for Interdisciplinary Research, Huntington, West Virginia (Y.X., P.M., M.H., T.W., L.C., Z.X.); University of Toledo College of Medicine and Life Sciences, Toledo, Ohio (J.X.X.); Joan C. Edwards School of Medicine at Marshall University, Huntington, West Virginia (J.I.S.); and Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, P. R. China, and Jiangsu Food and Pharmaceutical Science College, Huai'an, P. R. China (F.F.)
| | - Liquan Cai
- Marshall Institute for Interdisciplinary Research, Huntington, West Virginia (Y.X., P.M., M.H., T.W., L.C., Z.X.); University of Toledo College of Medicine and Life Sciences, Toledo, Ohio (J.X.X.); Joan C. Edwards School of Medicine at Marshall University, Huntington, West Virginia (J.I.S.); and Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, P. R. China, and Jiangsu Food and Pharmaceutical Science College, Huai'an, P. R. China (F.F.)
| | - Feng Feng
- Marshall Institute for Interdisciplinary Research, Huntington, West Virginia (Y.X., P.M., M.H., T.W., L.C., Z.X.); University of Toledo College of Medicine and Life Sciences, Toledo, Ohio (J.X.X.); Joan C. Edwards School of Medicine at Marshall University, Huntington, West Virginia (J.I.S.); and Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, P. R. China, and Jiangsu Food and Pharmaceutical Science College, Huai'an, P. R. China (F.F.)
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Huntington, West Virginia (Y.X., P.M., M.H., T.W., L.C., Z.X.); University of Toledo College of Medicine and Life Sciences, Toledo, Ohio (J.X.X.); Joan C. Edwards School of Medicine at Marshall University, Huntington, West Virginia (J.I.S.); and Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, P. R. China, and Jiangsu Food and Pharmaceutical Science College, Huai'an, P. R. China (F.F.)
| |
Collapse
|
17
|
Xu Y, Jiang X, Xu J, Qu W, Xie Z, Jiang RW, Feng F. A previously undescribed phenylethanoid glycoside from Callicarpa kwangtungensis Chun acts as an agonist of the Na/K-ATPase signal transduction pathway. PHYTOCHEMISTRY 2021; 181:112577. [PMID: 33190100 DOI: 10.1016/j.phytochem.2020.112577] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/23/2020] [Accepted: 10/24/2020] [Indexed: 06/11/2023]
Abstract
The new concept that Na/K-ATPase acts as a receptor prompted us to look for new ligands from Callicarpa kwangtungensis Chun. Using column chromatography, an undescribed phenethyl alcohol glycoside, callicarpanoside A, and an undescribed benzyl alcohol glycoside, callicarpanoside B, along with twelve known polyphenols were isolated from Callicarpa kwangtungensis Chun. All the isolated compounds were evaluated for their Na/K-ATPase (NKA) inhibitory activities. Using our NKA technology platform-based screening assay protocols, callicarpanoside B was identified as an undescribed Na/K-ATPase agonist. In particular, the newly identified benzyl alcohol glycoside was found to bind NKA and activate the receptor NKA/Src complex, resulting in the activation of protein kinase cascades. These cascades included extracellular signal-regulated kinases and protein kinase C epsilon, as well as NKA α1 endocytosis at nanomolar concentrations. Unlike the class of cardiotonic steroids, callicarpanoside B showed less inhibition of NKA activity and caused less cellular toxicity. Moreover, callicarpanoside B was found to bind NKA at a different site other than the cardiotonic steroids binding site. Thus, we have identified an undescribed NKA α1 agonist that may be used to enhance the physiological processes of NKA α1 signaling.
Collapse
Affiliation(s)
- Yunhui Xu
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV, 25701, United States
| | - Xueyang Jiang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China; Jiangsu Food and Pharmaceutical Science College, Huai'an, 223003, PR China
| | - Jian Xu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China; Jiangsu Food and Pharmaceutical Science College, Huai'an, 223003, PR China
| | - Wei Qu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China; Jiangsu Food and Pharmaceutical Science College, Huai'an, 223003, PR China
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV, 25701, United States
| | - Ren-Wang Jiang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, PR China; International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, College of Pharmacy, Jinan University, Guangzhou, 510632, PR China.
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China; Jiangsu Food and Pharmaceutical Science College, Huai'an, 223003, PR China.
| |
Collapse
|
18
|
Ouabain Promotes Gap Junctional Intercellular Communication in Cancer Cells. Int J Mol Sci 2020; 22:ijms22010358. [PMID: 33396341 PMCID: PMC7801950 DOI: 10.3390/ijms22010358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 12/17/2022] Open
Abstract
Gap junctions are molecular structures that allow communication between neighboring cells. It has been shown that gap junctional intercellular communication (GJIC) is notoriously reduced in cancer cells compared to their normal counterparts. Ouabain, a plant derived substance, widely known for its therapeutic properties on the heart, has been shown to play a role in several types of cancer, although its mechanism of action is not yet fully understood. Since we have previously shown that ouabain enhances GJIC in epithelial cells (MDCK), here we probed whether ouabain affects GJIC in a variety of cancer cell lines, including cervico-uterine (CasKi, SiHa and Hela), breast (MDA-MB-321 and MCF7), lung (A549), colon (SW480) and pancreas (HPAF-II). For this purpose, we conducted dye transfer assays to measure and compare GJIC in monolayers of cells with and without treatment with ouabain (0.1, 1, 10, 50 and 500 nM). We found that ouabain induces a statistically significant enhancement of GJIC in all of these cancer cell lines, albeit with distinct sensitivity. Additionally, we show that synthesis of new nucleotides or protein subunits is not required, and that Csrc, ErK1/2 and ROCK-Rho mediate the signaling mechanisms. These results may contribute to explaining how ouabain influences cancer.
Collapse
|
19
|
Qu X, Zhang Z, Hu W, Lou M, Zhai B, Mei S, Hu Z, Zhang L, Liu D, Liu Z, Chen J, Wang Y. Attenuation of the Na/K‑ATPase/Src/ROS amplification signaling pathway by astaxanthin ameliorates myocardial cell oxidative stress injury. Mol Med Rep 2020; 22:5125-5134. [PMID: 33173978 PMCID: PMC7646965 DOI: 10.3892/mmr.2020.11613] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/27/2020] [Indexed: 12/27/2022] Open
Abstract
The 3S, 3′S-ASTaxanthin (3S, 3′S-AST) isomer has strong antioxidant activity; however, its protective roles and potential mechanisms against oxidative stress damage in cardiomyocytes have not been investigated. Na+/K+-ATPase (NKA)/Src signal activation has an important role in increasing reactive oxygen species (ROS) production. The aim of the present study was to investigate the protective effects and mechanism of 3S, 3′S-AST on hydrogen peroxide (H2O2)-induced oxidative stress injury in H9c2 myocardial cells. The protective effects of 3S, 3′S-AST on H2O2-induced H9c2 cell injury was observed by measuring lactate dehydrogenase and creatine kinase myocardial band content, cell viability and nuclear morphology. The antioxidant effect was investigated by analyzing ROS accumulation and malondialdehyde, glutathione (GSH) peroxidase, GSH and glutathione reductase activity levels. The protein expression levels of Bax, Bcl-2, caspase-3 and cleaved caspase-3 were analyzed using western blotting to determine cardiomyocyte apoptosis. Western blot analysis of the phosphorylation levels of Src and Erk1/2 were also performed to elucidate the molecular mechanism involved. The results showed that 3S, 3′S-AST reduced the release of LDH and promoted cell viability, and attenuated ROS accumulation and cell apoptosis induced by H2O2. Furthermore, 3S, 3′S-AST also restored apoptosis-related Bax and Bcl-2 protein expression levels in H2O2-treated H9c2 cells. The phosphorylation levels of Src and Erk1/2 were significantly higher in the H2O2 treatment group, whereas 3S, 3′S-AST pretreatment significantly decreased the levels of phosphorylated (p)-Src and p-ERK1/2. The results provided evidence that 3S, 3′S-AST exhibited a cardioprotective effect against oxidative stress injury by attenuating NKA/Src/Erk1/2-modulated ROS amplification.
Collapse
Affiliation(s)
- Xuefeng Qu
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Zhouyi Zhang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Wenli Hu
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Minhan Lou
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Bingzhong Zhai
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Song Mei
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Zhihang Hu
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Lijing Zhang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Dongying Liu
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Zhen Liu
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Jianguo Chen
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Yin Wang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| |
Collapse
|
20
|
Petrushanko IY, Mitkevich VA, Makarov AA. Molecular Mechanisms of the Redox Regulation of the Na,K-ATPase. Biophysics (Nagoya-shi) 2020. [DOI: 10.1134/s0006350920050139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
21
|
Apical periodontitis induces changes on oxidative stress parameters and increases Na +/K +-ATPase activity in adult rats. Arch Oral Biol 2020; 118:104849. [PMID: 32847752 DOI: 10.1016/j.archoralbio.2020.104849] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/01/2020] [Accepted: 07/21/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Endodontic infection can cause systemic alterations. The involvement of oxidative stress (OS) and transmembrane enzymes compose the pathogenesis of various systemic diseases. However, the relation among apical periodontitis (AP), OS parameters, and Na+/K+-ATPase (NKA) pump was not reported in the literature. This study evaluated the AP influence on OS parameters and NKA activity in adult rats. METHODS Adult male Wistar rats (sixteen weeks old) were randomly assigned to two experimental groups: control (CT group; n = 8) and AP (AP group; n = 9), which was induced in the first right mandibular molar tooth. After 21 days of AP induction, mandibles were dissected for radiographic analysis. In addition, the heart, liver, pancreas, and kidney were collected for analysis of endogenous OS parameters and NKA activity. Data were analyzed by Student's T-test. Values of p < 0.05 were considered statistically significant. RESULTS AP presence increased reactive species (RS) generation only in the heart, while the other analyzed organs did not have this parameter modified. Heart and pancreas had a decreased endogenous antioxidant system (catalase activity and vitamin C levels), liver and kidney had an increased one. AP increased NKA activity in the heart, liver, and pancreas, but not in the kidney. CONCLUSION The modulation of both endogenous antioxidant defense system and NKA activity in vital organs suggested that alterations in the antioxidant status and cellular electrochemical gradient may be involved in the AP pathophysiology.
Collapse
|
22
|
Akkuratov EE, Westin L, Vazquez-Juarez E, de Marothy M, Melnikova AK, Blom H, Lindskog M, Brismar H, Aperia A. Ouabain Modulates the Functional Interaction Between Na,K-ATPase and NMDA Receptor. Mol Neurobiol 2020; 57:4018-4030. [PMID: 32651756 PMCID: PMC7467916 DOI: 10.1007/s12035-020-01984-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 06/08/2020] [Indexed: 12/18/2022]
Abstract
The N-methyl-D-aspartate (NMDA) receptor plays an essential role in glutamatergic transmission and synaptic plasticity and researchers are seeking for different modulators of NMDA receptor function. One possible mechanism for its regulation could be through adjacent membrane proteins. NMDA receptors coprecipitate with Na,K-ATPase, indicating a potential interaction of these two proteins. Ouabain, a mammalian cardiotonic steroid that specifically binds to Na,K-ATPase and affects its conformation, can protect from some toxic effects of NMDA receptor activation. Here we have examined whether NMDA receptor activity and downstream effects can be modulated by physiological ouabain concentrations. The spatial colocalization between NMDA receptors and the Na,K-ATPase catalytic subunits on dendrites of cultured rat hippocampal neurons was analyzed with super-resolution dSTORM microscopy. The functional interaction was analyzed with calcium imaging of single hippocampal neurons exposed to 10 μM NMDA in presence and absence of ouabain and by determination of the ouabain effect on NMDA receptor–dependent long-term potentiation. We show that NMDA receptors and the Na,K-ATPase catalytic subunits alpha1 and alpha3 exist in same protein complex and that ouabain in nanomolar concentration consistently reduces the calcium response to NMDA. Downregulation of the NMDA response is not associated with internalization of the receptor or with alterations in its state of Src phosphorylation. Ouabain in nanomolar concentration elicits a long-term potentiation response. Our findings suggest that ouabain binding to a fraction of Na,K-ATPase molecules that cluster with the NMDA receptors will, via a conformational effect on the NMDA receptors, cause moderate but consistent reduction of NMDA receptor response at synaptic activation.
Collapse
Affiliation(s)
- Evgeny E Akkuratov
- Science for Life Laboratory, Department of Applied Physics, Kungliga Tekniska Högskolan, Stockholm, Sweden
| | - Linda Westin
- Science for Life Laboratory, Department of Women's and Children's health, Karolinska Institutet, Stockholm, Sweden
| | - Erika Vazquez-Juarez
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Minttu de Marothy
- Science for Life Laboratory, Department of Women's and Children's health, Karolinska Institutet, Stockholm, Sweden
| | - Aleksandra K Melnikova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia, 119234
| | - Hans Blom
- Science for Life Laboratory, Department of Applied Physics, Kungliga Tekniska Högskolan, Stockholm, Sweden
| | - Maria Lindskog
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Hjalmar Brismar
- Science for Life Laboratory, Department of Applied Physics, Kungliga Tekniska Högskolan, Stockholm, Sweden.
| | - Anita Aperia
- Science for Life Laboratory, Department of Women's and Children's health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
23
|
Ju M, Ioannidou S, Munro P, Rämö O, Vihinen H, Jokitalo E, Shima DT. A Na,K-ATPase-Fodrin-Actin Membrane Cytoskeleton Complex is Required for Endothelial Fenestra Biogenesis. Cells 2020; 9:cells9061387. [PMID: 32503129 PMCID: PMC7349347 DOI: 10.3390/cells9061387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/30/2020] [Accepted: 05/30/2020] [Indexed: 01/07/2023] Open
Abstract
Fenestrae are transcellular plasma membrane pores that mediate blood–tissue exchange in specialised vascular endothelia. The composition and biogenesis of the fenestra remain enigmatic. We isolated and characterised the protein composition of large patches of fenestrated plasma membrane, termed sieve plates. Loss-of-function experiments demonstrated that two components of the sieve plate, moesin and annexin II, were positive and negative regulators of fenestra formation, respectively. Biochemical analyses showed that moesin is involved in the formation of an actin–fodrin submembrane cytoskeleton that was essential for fenestra formation. The link between the fodrin cytoskeleton and the plasma membrane involved the fenestral pore protein PV-1 and Na,K-ATPase, which is a key regulator of signalling during fenestra formation both in vitro and in vivo. These findings provide a conceptual framework for fenestra biogenesis, linking the dynamic changes in plasma membrane remodelling to the formation of a submembrane cytoskeletal signalling complex.
Collapse
Affiliation(s)
- Meihua Ju
- Translational Vision Research, UCL Institute of Ophthalmology, London EC1v 9EL, UK;
| | | | - Peter Munro
- Electron Microscopy Unit, UCL Institute of Ophthalmology, London EC1v 9EL, UK;
| | - Olli Rämö
- Cell and Molecular Biology Program, University of Helsinki, 00014 Helsinki, Finland; (O.R.); (E.J.)
| | - Helena Vihinen
- Electron Microscopy Unit, Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland;
| | - Eija Jokitalo
- Cell and Molecular Biology Program, University of Helsinki, 00014 Helsinki, Finland; (O.R.); (E.J.)
- Electron Microscopy Unit, Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland;
| | - David T. Shima
- Translational Vision Research, UCL Institute of Ophthalmology, London EC1v 9EL, UK;
- Correspondence: ; Tel.: +44-(0)7-932-042-570
| |
Collapse
|
24
|
The Na/K-ATPase α1 and c-Src form signaling complex under native condition: A crosslinking approach. Sci Rep 2020; 10:6006. [PMID: 32265464 PMCID: PMC7138855 DOI: 10.1038/s41598-020-61920-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 03/04/2020] [Indexed: 11/09/2022] Open
Abstract
The protein-protein interactions amongst the Na/K-ATPase α1 subunit, c-Src, and caveolin-1 (cav-1) are essential for the Na/K-ATPase signaling functions. However, there are arguments concerning the interaction model. The present study aims to clarify the interactions amongst the endogenous native proteins in live cells under native resting condition. Under native condition, Blue Native-PAGE and Blue Native-PAGE/SDS-PAGE 2D analyses demonstrated co-existence of the α1 subunit and c-Src in same protein complex, as well as a direct interaction between the α1 subunit and c-Src. By comparison of cleavable and non-cleavable cysteine-cysteine crosslinked samples, capillary immunoblotting analysis demonstrated that depletion of Src kinase family members (c-Src, Yes, and Fyn) or cav-1 clearly reduced the interactions of the α1 subunit with proteins, but depletion of cav-1 did not affect the interaction of c-Src with the α1 subunit. The data indicated that there are direct interactions between the α1 subunit and c-Src as well as between the α1 subunit and cav-1, but argued about the interaction between c-Src and cav-1 under the condition. Furthermore, the data also indicated the existence of different protein complexes containing the α1 subunit and c-Src, which might have different signaling functions.
Collapse
|
25
|
Liu J, Nie Y, Chaudhry M, Bai F, Chuang J, Sodhi K, Shapiro JI. The Redox-Sensitive Na/K-ATPase Signaling in Uremic Cardiomyopathy. Int J Mol Sci 2020; 21:ijms21041256. [PMID: 32069992 PMCID: PMC7072896 DOI: 10.3390/ijms21041256] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 02/07/2023] Open
Abstract
In recent years, Na/K-ATPase signaling has been implicated in different physiological and pathophysiological conditions, including cardiac hypertrophy and uremic cardiomyopathy. Cardiotonic steroids (CTS), specific ligands of Na/K-ATPase, regulate its enzymatic activity (at higher concentrations) and signaling function (at lower concentrations without significantly affecting its enzymatic activity) and increase reactive oxygen species (ROS) generation. On the other hand, an increase in ROS alone also regulates the Na/K-ATPase enzymatic activity and signaling function. We termed this phenomenon the Na/K-ATPase-mediated oxidant-amplification loop, in which oxidative stress regulates both the Na/K-ATPase activity and signaling. Most recently, we also demonstrated that this amplification loop is involved in the development of uremic cardiomyopathy. This review aims to evaluate the redox-sensitive Na/K-ATPase-mediated oxidant amplification loop and uremic cardiomyopathy.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (Y.N.); (M.C.); (F.B.)
- Correspondence:
| | - Ying Nie
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (Y.N.); (M.C.); (F.B.)
| | - Muhammad Chaudhry
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (Y.N.); (M.C.); (F.B.)
| | - Fang Bai
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (Y.N.); (M.C.); (F.B.)
| | - Justin Chuang
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (J.C.); (K.S.); (J.I.S.)
| | - Komal Sodhi
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (J.C.); (K.S.); (J.I.S.)
| | - Joseph I. Shapiro
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (J.C.); (K.S.); (J.I.S.)
| |
Collapse
|
26
|
Blaustein MP, Hamlyn JM. Ouabain, endogenous ouabain and ouabain-like factors: The Na + pump/ouabain receptor, its linkage to NCX, and its myriad functions. Cell Calcium 2020; 86:102159. [PMID: 31986323 DOI: 10.1016/j.ceca.2020.102159] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/01/2020] [Accepted: 01/03/2020] [Indexed: 12/12/2022]
Abstract
In this brief review we discuss some aspects of the Na+ pump and its roles in mediating the effects of ouabain and endogenous ouabain (EO): i) in regulating the cytosolic Ca2+ concentration ([Ca2+]CYT) via Na/Ca exchange (NCX), and ii) in activating a number of protein kinase (PK) signaling cascades that control a myriad of cell functions. Importantly, [Ca2+]CYT and the other signaling pathways intersect at numerous points because of the influence of Ca2+ and calmodulin in modulating some steps in those other pathways. While both mechanisms operate in virtually all cells and tissues, this article focuses primarily on their functions in the cardiovascular system, the central nervous system (CNS) and the kidneys.
Collapse
Affiliation(s)
- Mordecai P Blaustein
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - John M Hamlyn
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
27
|
Influence of Endogenous Cardiac Glycosides, Digoxin, and Marinobufagenin in the Physiology of Epithelial Cells. Cardiol Res Pract 2019; 2019:8646787. [PMID: 32089875 PMCID: PMC7024086 DOI: 10.1155/2019/8646787] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/20/2019] [Accepted: 11/15/2019] [Indexed: 12/14/2022] Open
Abstract
Cardiac glycosides are a group of compounds widely known for their action in cardiac tissue, some of which have been found to be endogenously produced (ECG). We have previously studied the effect of ouabain, an endogenous cardiac glycoside, on the physiology of epithelial cells, and we have shown that in concentrations in the nanomolar range, it affects key properties of epithelial cells, such as tight junction, apical basolateral polarization, gap junctional intercellular communication (GJIC), and adherent junctions. In this work, we study the influence of digoxin and marinobufagenin, two other endogenously expressed cardiac glycosides, on GJIC as well as the degree of transepithelial tightness due to tight junction integrity (TJ). We evaluated GJIC by dye transfer assays and tight junction integrity by transepithelial electrical resistance (TER) measurements, as well as immunohistochemistry and western blot assays of expression of claudins 2 and 4. We found that both digoxin and marinobufagenin improve GJIC and significantly enhance the tightness of the tight junctions, as evaluated from TER measurements. Immunofluorescence assays show that both compounds promote enhanced basolateral localization of claudin-4 but not claudin 2, while densitometric analysis of western blot assays indicate a significantly increased expression of claudin 4. These changes, induced by digoxin and marinobufagenin on GJIC and TER, were not observed on MDCK-R, a modified MDCK cell line that has a genetically induced insensitive α1 subunit, indicating that Na-K-ATPase acts as a receptor mediating the actions of both ECG. Plus, the fact that the effect of both cardiac glycosides was suppressed by incubation with PP2, an inhibitor of c-Src kinase, PD98059, an inhibitor of mitogen extracellular kinase-1 and Y-27632, a selective inhibitor of ROCK, and a Rho-associated protein kinase, indicate altogether that the signaling pathways involved include c-Src and ERK1/2, as well as Rho-ROCK. These results widen and strengthen our general hypothesis that a very important physiological role of ECG is the control of the epithelial phenotype and the regulation of cell-cell contacts.
Collapse
|
28
|
Pavlovic D. Endogenous cardiotonic steroids and cardiovascular disease, where to next? Cell Calcium 2019; 86:102156. [PMID: 31896530 PMCID: PMC7031694 DOI: 10.1016/j.ceca.2019.102156] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 12/24/2019] [Accepted: 12/24/2019] [Indexed: 11/18/2022]
Abstract
Ever since British Physician William Withering first described the use of foxglove extract for treatment of patients with congestive heart failure in 1785, cardiotonic steroids have been used clinically to treat heart failure and more recently atrial fibrillation. Due to their ability to bind and inhibit the ubiquitous transport enzyme sodium potassium pump, thus regulating intracellular Na+ concentration in every living cell, they are also an essential tool for research into the sodium potassium pump structure and function. Exogenous CTS have been clearly demonstrated to affect cardiovascular system through modulation of vagal tone, cardiac contraction (via ionic changes) and altered natriuresis. Reports of a number of endogenous CTS, since the 1980s, have intensified research into their physiologic and pathophysiologic roles and opened up novel therapeutic targets. Substantive evidence pointing to the role of endogenous ouabain and marinobufagenin, the two most prominent CTS, in development of cardiovascular disease has accumulated. Nevertheless, their presence, structure, biosynthesis pathways and even mechanism of action remain unclear or controversial. In this review the current state-of-the-art, the controversies and the remaining questions surrounding the role of endogenous cardiotonic steroids in health and disease are discussed.
Collapse
Affiliation(s)
- Davor Pavlovic
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
29
|
Meade JC. P-type transport ATPases in Leishmania and Trypanosoma. ACTA ACUST UNITED AC 2019; 26:69. [PMID: 31782726 PMCID: PMC6884021 DOI: 10.1051/parasite/2019069] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 11/12/2019] [Indexed: 01/12/2023]
Abstract
P-type ATPases are critical to the maintenance and regulation of cellular ion homeostasis and membrane lipid asymmetry due to their ability to move ions and phospholipids against a concentration gradient by utilizing the energy of ATP hydrolysis. P-type ATPases are particularly relevant in human pathogenic trypanosomatids which are exposed to abrupt and dramatic changes in their external environment during their life cycles. This review describes the complete inventory of ion-motive, P-type ATPase genes in the human pathogenic Trypanosomatidae; eight Leishmania species (L. aethiopica, L. braziliensis, L. donovani, L. infantum, L. major, L. mexicana, L. panamensis, L. tropica), Trypanosoma cruzi and three Trypanosoma brucei subspecies (Trypanosoma brucei brucei TREU927, Trypanosoma brucei Lister strain 427, Trypanosoma brucei gambiense DAL972). The P-type ATPase complement in these trypanosomatids includes the P1B (metal pumps), P2A (SERCA, sarcoplasmic-endoplasmic reticulum calcium ATPases), P2B (PMCA, plasma membrane calcium ATPases), P2D (Na+ pumps), P3A (H+ pumps), P4 (aminophospholipid translocators), and P5B (no assigned specificity) subfamilies. These subfamilies represent the P-type ATPase transport functions necessary for survival in the Trypanosomatidae as P-type ATPases for each of these seven subfamilies are found in all Leishmania and Trypanosoma species included in this analysis. These P-type ATPase subfamilies are correlated with current molecular and biochemical knowledge of their function in trypanosomatid growth, adaptation, infectivity, and survival.
Collapse
Affiliation(s)
- John C Meade
- Department of Microbiology and Immunology, School of Medicine, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| |
Collapse
|
30
|
Hanafusa K, Hayashi N. The Flot2 component of the lipid raft changes localization during neural differentiation of P19C6 cells. BMC Mol Cell Biol 2019; 20:38. [PMID: 31455216 PMCID: PMC6712619 DOI: 10.1186/s12860-019-0225-0] [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: 08/14/2019] [Accepted: 08/22/2019] [Indexed: 12/18/2022] Open
Abstract
Background Flotillin-2 (Flot2) is a lipid raft scaffold protein that is thought to be related to neural differentiation. Flot2 is phosphorylated by Fyn, a Src kinase, and causes raft-dependent endocytosis; however, the exact role of Flot2 in neural differentiation remains unclear. To reveal the roles of lipid raft-associated proteins during neural differentiation, we tried to analyze the expression and localization. Results In this study, we found that the expression levels of the Flot2 and Fyn proteins increased in whole-cell lysates of P19C6 cells after neural differentiation. In addition, sucrose density fractionation and immunofluorescence experiments revealed an increase in the localization of Flot2 and Fyn to lipid rafts after neural differentiation. We also found that Fyn partially colocalized with Flot2 lipid rafts in neural cells. Conclusion The observed distribution of Fyn and level of inactivated Fyn and/or c-Src in detergent–resistant membrane (DRM) fractions suggests that the amount of activated Fyn might increase in DRM fractions after neural differentiation. Overall these findings suggest that Flot2 lipid rafts are associated with Fyn, and that Fyn phosphorylates Flot2 during neural differentiation of P19C6 cells. Electronic supplementary material The online version of this article (10.1186/s12860-019-0225-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kei Hanafusa
- School of Life Science and Technology, Tokyo Institute of Technology, Meguro-ku, Tokyo, Japan
| | - Nobuhiro Hayashi
- School of Life Science and Technology, Tokyo Institute of Technology, Meguro-ku, Tokyo, Japan.
| |
Collapse
|
31
|
Panizza E, Zhang L, Fontana JM, Hamada K, Svensson D, Akkuratov EE, Scott L, Mikoshiba K, Brismar H, Lehtiö J, Aperia A. Ouabain-regulated phosphoproteome reveals molecular mechanisms for Na +, K +-ATPase control of cell adhesion, proliferation, and survival. FASEB J 2019; 33:10193-10206. [PMID: 31199885 PMCID: PMC6704450 DOI: 10.1096/fj.201900445r] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ion pump Na+, K+-ATPase (NKA) is a receptor for the cardiotonic steroid ouabain. Subsaturating concentration of ouabain triggers intracellular calcium oscillations, stimulates cell proliferation and adhesion, and protects from apoptosis. However, it is controversial whether ouabain-bound NKA is considered a signal transducer. To address this question, we performed a global analysis of protein phosphorylation in COS-7 cells, identifying 2580 regulated phosphorylation events on 1242 proteins upon 10- and 20-min treatment with ouabain. Regulated phosphorylated proteins include the inositol triphosphate receptor and stromal interaction molecule, which are essential for initiating calcium oscillations. Hierarchical clustering revealed that ouabain triggers a structured phosphorylation response that occurs in a well-defined, time-dependent manner and affects specific cellular processes, including cell proliferation and cell-cell junctions. We additionally identify regulation of the phosphorylation of several calcium and calmodulin-dependent protein kinases (CAMKs), including 2 sites of CAMK type II-γ (CAMK2G), a protein known to regulate apoptosis. To verify the significance of this result, CAMK2G was knocked down in primary kidney cells. CAMK2G knockdown impaired ouabain-dependent protection from apoptosis upon treatment with high glucose or serum deprivation. In conclusion, we establish NKA as the coordinator of a broad, tightly regulated phosphorylation response in cells and define CAMK2G as a downstream effector of NKA.-Panizza, E., Zhang, L., Fontana, J. M., Hamada, K., Svensson, D., Akkuratov, E. E., Scott, L., Mikoshiba, K., Brismar, H., Lehtiö, J., Aperia, A. Ouabain-regulated phosphoproteome reveals molecular mechanisms for Na+, K+-ATPase control of cell adhesion, proliferation, and survival.
Collapse
Affiliation(s)
- Elena Panizza
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Solna, Sweden
| | - Liang Zhang
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Jacopo Maria Fontana
- Department of Applied Physics, Science for Life Laboratory, Royal Institute of Technology, Stockholm, Sweden
| | - Kozo Hamada
- Laboratory for Developmental Neurobiology, Brain Science Institute, Riken, Saitama, Japan
| | - Daniel Svensson
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Evgeny E Akkuratov
- Department of Applied Physics, Science for Life Laboratory, Royal Institute of Technology, Stockholm, Sweden
| | - Lena Scott
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, Brain Science Institute, Riken, Saitama, Japan
| | - Hjalmar Brismar
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden.,Department of Applied Physics, Science for Life Laboratory, Royal Institute of Technology, Stockholm, Sweden
| | - Janne Lehtiö
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Solna, Sweden
| | - Anita Aperia
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
32
|
Characterization, quantitation, similarity evaluation and combination with Na+,K+-ATPase of cardiac glycosides from Streblus asper. Bioorg Chem 2019; 87:265-275. [DOI: 10.1016/j.bioorg.2019.03.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/09/2019] [Accepted: 03/16/2019] [Indexed: 12/15/2022]
|
33
|
The Adipocyte Na/K-ATPase Oxidant Amplification Loop is the Central Regulator of Western Diet-Induced Obesity and Associated Comorbidities. Sci Rep 2019; 9:7927. [PMID: 31138824 PMCID: PMC6538745 DOI: 10.1038/s41598-019-44350-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 05/10/2019] [Indexed: 12/29/2022] Open
Abstract
Obesity has become a worldwide epidemic. We have previously reported that systemic administration of pNaKtide which targets the Na/K-ATPase oxidant amplification loop (NKAL) was able to decrease systemic oxidative stress and adiposity in mice fed a high fat and fructose supplemented western diet (WD). As adipocytes are believed to play a central role in the development of obesity and its related comorbidities, we examined whether lentiviral-mediated adipocyte-specific expression of NaKtide, a peptide derived from the N domain of the alpha1 Na/K-ATPase subunit, could ameliorate the effects of the WD. C57BL6 mice were fed a WD, which activated Na/K-ATPase signaling in the adipocytes and induced an obese phenotype and caused an increase in plasma levels of leptin, IL-6 and TNFα. WD also decreased locomotor activity, expression of the D2 receptor and tyrosine hydroxylase in brain tissue, while markers of neurodegeneration and neuronal apoptosis were increased following the WD. Selective adipocyte expression of NaKtide in these mice fed a WD attenuated all of these changes including the brain biochemical alterations and behavioral adaptations. These data suggest that adipocyte derived cytokines play an essential role in the development of obesity induced by a WD and that targeting the adipocyte NKAL loop may serve as an effective therapeutic strategy.
Collapse
|
34
|
Litan A, Li Z, Tokhtaeva E, Kelly P, Vagin O, Langhans SA. A Functional Interaction Between Na,K-ATPase β 2-Subunit/AMOG and NF2/Merlin Regulates Growth Factor Signaling in Cerebellar Granule Cells. Mol Neurobiol 2019; 56:7557-7571. [PMID: 31062247 DOI: 10.1007/s12035-019-1592-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 04/02/2019] [Indexed: 10/26/2022]
Abstract
The Na,K-ATPase, consisting of a catalytic α-subunit and a regulatory β-subunit, is a ubiquitously expressed ion pump that carries out the transport of Na+ and K+ across the plasma membranes of most animal cells. In addition to its pump function, Na,K-ATPase serves as a signaling scaffold and a cell adhesion molecule. Of the three β-subunit isoforms, β1 is found in almost all tissues, while β2 expression is mostly restricted to brain and muscle. In cerebellar granule cells, the β2-subunit, also known as adhesion molecule on glia (AMOG), has been linked to neuron-astrocyte adhesion and granule cell migration, suggesting its role in cerebellar development. Nevertheless, little is known about molecular pathways that link the β2-subunit to its cellular functions. Using cerebellar granule precursor cells, we found that the β2-subunit, but not the β1-subunit, negatively regulates the expression of a key activator of the Hippo/YAP signaling pathway, Merlin/neurofibromin-2 (NF2). The knockdown of the β2-subunit resulted in increased Merlin/NF2 expression and affected downstream targets of Hippo signaling, i.e., increased YAP phosphorylation and decreased expression of N-Ras. Further, the β2-subunit knockdown altered the kinetics of epidermal growth factor receptor (EGFR) signaling in a Merlin-dependent mode and impaired EGF-induced reorganization of the actin cytoskeleton. Therefore, our studies for the first time provide a functional link between the Na,K-ATPase β2-subunit and Merlin/NF2 and suggest a role for the β2-subunit in regulating cytoskeletal dynamics and Hippo/YAP signaling during neuronal differentiation.
Collapse
Affiliation(s)
- Alisa Litan
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, DuPont Experimental Station, Bldg 400, #4414, 200 Powder Mill Road, Wilmington, DE, 19803, USA.,Biological Sciences Graduate Program, University of Delaware, Newark, DE, 19716, USA
| | - Zhiqin Li
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, DuPont Experimental Station, Bldg 400, #4414, 200 Powder Mill Road, Wilmington, DE, 19803, USA
| | - Elmira Tokhtaeva
- David Geffen School of Medicine, University of California, Los Angeles, and VA Greater Los Angeles Health Care System, Los Angeles, CA, 90073, USA
| | - Patience Kelly
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, DuPont Experimental Station, Bldg 400, #4414, 200 Powder Mill Road, Wilmington, DE, 19803, USA.,Biological Sciences Graduate Program, University of Delaware, Newark, DE, 19716, USA
| | - Olga Vagin
- David Geffen School of Medicine, University of California, Los Angeles, and VA Greater Los Angeles Health Care System, Los Angeles, CA, 90073, USA
| | - Sigrid A Langhans
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, DuPont Experimental Station, Bldg 400, #4414, 200 Powder Mill Road, Wilmington, DE, 19803, USA.
| |
Collapse
|
35
|
Chen L, Jiang P, Li J, Xie Z, Xu Y, Qu W, Feng F, Liu W. Periplocin promotes wound healing through the activation of Src/ERK and PI3K/Akt pathways mediated by Na/K-ATPase. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 57:72-83. [PMID: 30668325 DOI: 10.1016/j.phymed.2018.12.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/01/2018] [Accepted: 12/10/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Periploca forrestii(PF) is mainly utilized for treatment of arthritis and traumatic injury historically. We had previously demonstrated that a fraction rich in cardiotonic steroids isolated from PF had the potential to facilitate wound healing. However, the exact material basis and mechanism of action responsible for wound healing is still unclear. Periplocin(PP) is the highest level of cardiotonic steroid included in PF. The present study aims to evaluate the efficacy of periplocin on wound healing systematically in vitro and in vivo. MATERIALS AND METHODS The L929 proliferation was determined by both MTT and EdU assay. Cell migration was tested by both scratch and transwell assay. The total amount of soluble collagen was assessed using a Sircol Collagen Assay Kit. The wound healing activity was evaluated in vivo using the excision rat models. Histopathology of the wounded skin on day 9 was studied via hematoxylin and eosin staining (HE) for general morphological observations and masson's trichrome staining for collagen deposition, respectively. The alteration in Src/ERK and PI3K/Akt pathways mediated by Na/K-ATPase was determined by western blot after the treatment with periplocin. The interaction between Na/K-ATPase and Src was tested by immunoprecipitation and immunostaining analysis. RESULTS The results revealed that periplocin could significantly boost proliferation, migration and stimulate collagen production in fibroblast L929 cells, which is dependent on activation of Src/ERK and PI3K/Akt pathways mediated by Na/K-ATPase, and thus promoting wound healing. Indeed, inhibition of Na/K-ATPase/Src complex receptor by Src specific inhibitor or knocking down the Na/K-ATPase expression would abolish the subsequent activation of Src/ERK and PI3K/Akt pathways and attenuate periplocin-induced beneficial effects on wound healing. Additionally, the wound healing activity is also confirmed in a rat excisional wound model as evidenced by increased rate of wound closure, reepithelization, formation of granulation tissue and collagen accumulation. CONCLUSIONS Collectively, we lay the rationale for traditional usage for traumatic injury, suggesting that periplocin and periploca forrestii is a promising candidate for management of chronic wounds.
Collapse
Affiliation(s)
- Lei Chen
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
| | - Pan Jiang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Jinsong Li
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
| | - Zijian Xie
- Department of Physiology and Pharmacology, Mail Stop 1008, College of Medicine, University of Toledo, 3000 Arlington Avenue, Toledo, OH 43614, USA
| | - Yunhui Xu
- Department of Physiology and Pharmacology, Mail Stop 1008, College of Medicine, University of Toledo, 3000 Arlington Avenue, Toledo, OH 43614, USA
| | - Wei Qu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China; Jiangsu Food & Pharmaceutical Science College, 4 Meicheng Donglu, Huaian 223003, China.
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
36
|
Chen L, Li J, Ke X, Sun C, Huang X, Jiang P, Feng F, Liu W, Zhang J. Chemical profiling and the potential active constituents responsible for wound healing in Periploca forrestii Schltr. JOURNAL OF ETHNOPHARMACOLOGY 2018; 224:230-241. [PMID: 29680302 DOI: 10.1016/j.jep.2018.04.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 03/28/2018] [Accepted: 04/17/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Periploca forrestii Schltr. is a popular folk medicine in china, commonly prescribed for the treatment of rheumatoid arthritis and wounds. The present research aimed to evaluate the effects of HLG on wound healing and reveal the potential active constituents. MATERIALS AND METHODS The wound healing activity was assessed by proliferation of fibroblast, migration and collagen production using L929 cells. A reliable HPLC-Q-TOF-MS/MS method was constructed for the systematic identification and characterization of main components in HLG. For further clarifying the potential active ingredients responsible for wound healing, total extract was separated by D101 macroporous resin. The fraction with strongest potency on wound healing was screened out by comparing with total extract. Finally, a new quantitative method was developed for determination of four typical cardiac glycosides in HLG by LC-MS. RESULTS The results showed that the total extract significantly promoted proliferation of fibroblast L929 up to 168% at 50 μg/ml. It also notably enhanced L929 migration on day 2 up to 56% and stimulated collagen release (96.1 μg/ml) at 50 μg/ml. A total of 38 compounds were identified or tentatively characterized by HPLC-Q-TOF-MS/MS based on reference substances or literatures. The separation by D101 macroporous adsorption resin led to the identification of 65 ethanol eluate as the most effective fraction. The data suggested that it could markedly promote L929 growth (174% of control), accelerate wound contraction (63% on day 2) and stimulate collagen generation (103.7 μg/ml) at 50 μg/ml, all of which were comparable to those of total extract. Interestingly, the HPLC-Q-TOF-MS/MS analysis revealed that the 65 ethanol fraction was mainly composed of cardiac glycosides. Finally, the new quantitative method was successfully utilized for detection of four typical cardiac glycosides in HLG, showing good performance in terms of analytical methodology. CONCLUSION The present study identified the cardiac glycosides as potential active constituents associated with wound healing and might afford a chemical foundation for preparation development of crude drug and quality evaluation of relevant products.
Collapse
Affiliation(s)
- Lei Chen
- Department of Pharmaceutical Analysis, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Jinsong Li
- Department of Pharmaceutical Analysis, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Xuan Ke
- Department of Pharmacology, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Cailing Sun
- School of Traditional Chinese pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Xiaoxian Huang
- School of Traditional Chinese pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Pan Jiang
- School of Traditional Chinese pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Feng Feng
- School of Traditional Chinese pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China.
| | - Jie Zhang
- School of Traditional Chinese pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China.
| |
Collapse
|
37
|
Telocinobufagin and Marinobufagin Produce Different Effects in LLC-PK1 Cells: A Case of Functional Selectivity of Bufadienolides. Int J Mol Sci 2018; 19:ijms19092769. [PMID: 30223494 PMCID: PMC6163863 DOI: 10.3390/ijms19092769] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/09/2018] [Accepted: 09/13/2018] [Indexed: 12/24/2022] Open
Abstract
Bufadienolides are cardiotonic steroids (CTS) identified in mammals. Besides Na+/K+-ATPase inhibition, they activate signal transduction via protein–protein interactions. Diversity of endogenous bufadienolides and mechanisms of action may indicate the presence of functional selectivity and unique cellular outcomes. We evaluated whether the bufadienolides telocinobufagin and marinobufagin induce changes in proliferation or viability of pig kidney (LLC-PK1) cells and the mechanisms involved in these changes. In some experiments, ouabain was used as a positive control. CTS exhibited an inhibitory IC50 of 0.20 (telocinobufagin), 0.14 (ouabain), and 3.40 μM (marinobufagin) for pig kidney Na+/K+-ATPase activity and concentrations that barely inhibited it were tested in LLC-PK1 cells. CTS induced rapid ERK1/2 phosphorylation, but corresponding proliferative response was observed for marinobufagin and ouabain instead of telocinobufagin. Telocinobufagin increased Bax:Bcl-2 expression ratio, sub-G0 cell cycle phase and pyknotic nuclei, indicating apoptosis. Src and MEK1/2 inhibitors blunted marinobufagin but not telocinobufagin effect, which was also not mediated by p38, JNK1/2, and PI3K. However, BIO, a GSK-3β inhibitor, reduced proliferation and, as telocinobufagin, phosphorylated GSK-3β at inhibitory Ser9. Combination of both drugs resulted in synergistic antiproliferative effect. Wnt reporter activity assay showed that telocinobufagin impaired Wnt/β-catenin pathway by acting upstream to β-catenin stabilization. Our findings support that mammalian endogenous bufadienolides may exhibit functional selectivity.
Collapse
|
38
|
The Na/K-ATPase Signaling: From Specific Ligands to General Reactive Oxygen Species. Int J Mol Sci 2018; 19:ijms19092600. [PMID: 30200500 PMCID: PMC6163532 DOI: 10.3390/ijms19092600] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/10/2018] [Accepted: 08/28/2018] [Indexed: 12/16/2022] Open
Abstract
The signaling function of the Na/K-ATPase has been established for 20 years and is widely accepted in the field, with many excellent reports and reviews not cited here. Even though there is debate about the underlying mechanism, the signaling function is unquestioned. This short review looks back at the evolution of Na/K-ATPase signaling, from stimulation by cardiotonic steroids (also known as digitalis-like substances) as specific ligands to stimulation by reactive oxygen species (ROS) in general. The interplay of cardiotonic steroids and ROS in Na/K-ATPase signaling forms a positive-feedback oxidant amplification loop that has been implicated in some pathophysiological conditions.
Collapse
|
39
|
DR region of Na +-K +-ATPase is a new target to protect heart against oxidative injury. Sci Rep 2018; 8:13100. [PMID: 30166619 PMCID: PMC6117330 DOI: 10.1038/s41598-018-31460-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 08/20/2018] [Indexed: 01/11/2023] Open
Abstract
Previous studies have shown that the activity and expression of Na+/K+-ATPase (NKA) are down-regulated in the failing hearts, and that an antibody against the DR-region of NKA (DR-Ab) can stimulate its activity. The present study was designed to investigate the beneficial effects of this antibody against cardiac injury and the underlying mechanisms. We found that DR-Ab improved cardiac function, alleviated cardiac hypertrophy and reduced oxidative stress in isoproterenol-treated mice. In AC16 human cardiomyocytes, DR-Ab increased cell viability and attenuated apoptosis under oxidative stress. Corresponding to the observation of reduced NKA activity, NKA abundance on plasma membrane was lowered during oxidative stress. Suppressed activity of protein phosphatase 2 A (PP2A) was responsible for the loss of membrane NKA due to the increased phosphorylation of key serine residues that trigger endocytosis. Incubation with DR-Ab restored PP2A activity and stabilized NKA expression on the plasma membrane. Inhibitors of PP2A abolished the protective effect of DR-Ab against oxidative stress. In summary, our data indicate that loss of membrane NKA may contribute to cardiac pathologies in heart failure. DR-Ab, by stabilizing membrane NKA, protects cardiomyocytes against oxidative injury and improves cardiac function in the failing hearts, suggesting a novel approach to treat heart failure.
Collapse
|
40
|
Banerjee M, Cui X, Li Z, Yu H, Cai L, Jia X, He D, Wang C, Gao T, Xie Z. Na/K-ATPase Y260 Phosphorylation-mediated Src Regulation in Control of Aerobic Glycolysis and Tumor Growth. Sci Rep 2018; 8:12322. [PMID: 30120256 PMCID: PMC6098021 DOI: 10.1038/s41598-018-29995-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 07/23/2018] [Indexed: 12/22/2022] Open
Abstract
We report here the identification of α1 Na/K-ATPase as a major regulator of the proto-oncogene Src kinase and the role of this regulation in control of Warburg effect and tumor growth. Specifically, we discovered Y260 in α1 Na/K-ATPase as a Src-specific phosphorylation and binding site and that Y260 phosphorylation is required for Src-mediated signal transduction in response to a number of stimuli including EGF. As such, it enables a dynamic control of aerobic glycolysis. However, such regulation appears to be lost or attenuated in human cancers as the expression of Na/K-ATPase α1 was significantly decreased in prostate, breast and kidney cancers, and further reduced in corresponding metastatic lesions in patient samples. Consistently, knockdown of α1 Na/K-ATPase led to a further increase in lactate production and the growth of tumor xenograft. These findings suggest that α1 Na/K-ATPase works as a tumor suppressor and that a loss of Na/K-ATPase-mediated Src regulation may lead to Warburg phenotype in cancer.
Collapse
Affiliation(s)
- Moumita Banerjee
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, 25703, USA
| | - Xiaoyu Cui
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, 25703, USA
| | - Zhichuan Li
- Department of Physiology and Pharmacology and Medicine, University of Toledo College of Medicine, Toledo, Ohio, 43614, USA
| | - Hui Yu
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, 25703, USA
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Liquan Cai
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, 25703, USA
| | - Xuelian Jia
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, 25703, USA
| | - Daheng He
- Department of Cancer Biostatistics, Markey Cancer Research Center, University of Kentucky, Lexington, Kentucky, 40536, USA
| | - Chi Wang
- Department of Cancer Biostatistics, Markey Cancer Research Center, University of Kentucky, Lexington, Kentucky, 40536, USA
| | - Tianyan Gao
- Department of Molecular and Cellular Biochemistry, Markey Cancer Research Center, University of Kentucky, Lexington, Kentucky, 40536, USA
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research (MIIR), Marshall University, Huntington, West Virginia, 25703, USA.
| |
Collapse
|
41
|
Khajah MA, Mathew PM, Luqmani YA. Na+/K+ ATPase activity promotes invasion of endocrine resistant breast cancer cells. PLoS One 2018; 13:e0193779. [PMID: 29590154 PMCID: PMC5874017 DOI: 10.1371/journal.pone.0193779] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 02/17/2018] [Indexed: 01/27/2023] Open
Abstract
Background The Na+/K+-ATPase (NKP) is an important ion transporter also involved in signal transduction. Its expression profile is altered in various tumours including that of the breast. We studied the effect of inhibiting NKP activity in non-tumorigenic breast cell line and in estrogen receptor positive and negative breast cancer cells. Methods Expression and localization of NKP and downstream signaling molecules were determined by RT-PCR, western blotting and immunofluorescence. Cell proliferation, apoptosis and cell cycle stage were determined using MTT, annexin V and flow cytometry. Cell motility and invasion were determined using wound healing and matrigel assays. Total matrix metalloproteinase (MMP) was determined by a fluorescence-based assay. Results NKP was mainly localized on the cell membrane. Its baseline expression and activity were enhanced in breast cancer compared to the non-tumorigenic breast cell line. Ouabain and 3,4,5,6-tetrahydroxyxanthone (TTX) treatment significantly inhibited NKP activity, which significantly reduced cell proliferation, motility, invasion and pH-induced membrane blebbing. EGF stimulation induced internalization of NKP from the cell membrane to the cytoplasm. Ouabain inhibited EGF-induced phosphorylation of Rac/cdc42, profillin, ERK1/2 and P70S6K. Conclusions The NKP may offer a novel therapeutic target in breast cancer patients who have developed metastasis, aiming to improve therapeutic outcomes and enhance survival rate.
Collapse
|
42
|
Feng X, Wang C, Xu Y, Turley J, Xie Z, Pierre SV, Hao J. Topical Digitoxigenin for Wound Healing: A Feasibility Study. Bioengineering (Basel) 2018; 5:bioengineering5010021. [PMID: 29510580 PMCID: PMC5874887 DOI: 10.3390/bioengineering5010021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 02/24/2018] [Accepted: 03/02/2018] [Indexed: 01/02/2023] Open
Abstract
(1) Background: Cardiotonic steroids have been found to stimulate collagen synthesis and might be potential wound healing therapeutics. The objective of this study was to evaluate the feasibility of digitoxigenin and its topical formulation for wound healing; (2) Methods: In the in vitro study, the human dermal fibroblast cells were treated with digitoxigenin and collagen synthesis was assessed. In the in vivo study, digitoxigenin was applied to excisional full-thickness wounds in rats immediately after wounding and remained for three days, and wound open was evaluated over 10 days. A digitoxigenin formulation for topical administration was prepared, and the in vitro release and in vivo wound healing effect were investigated; (3) Results: The expression of procollagen in human dermal fibroblast was significantly increased with the exposure to 0.1 nM digitoxigenin. Topical application of digitoxigenin in olive oil or alginate solution for three days significantly decreased the wound open in rats. Similarly, topical administration of the developed digitoxigenin formulation for three days also significantly increased wound healing. No wound healing effects were observed at days 7 and 10 after wounding when digitoxigenin was not applied; and, (4) Conclusions: It was possible to deliver digitoxigenin using the developed formulation. However, the wound healing effect of digitoxigenin and its mechanisms need to be further investigated in future studies.
Collapse
Affiliation(s)
- Xinchi Feng
- Department of Pharmaceutical Science and Research, School of Pharmacy, Marshall University, One John Marshall Drive, Huntington, WV 25755, USA.
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| | - Cuifen Wang
- Department of Pharmaceutical Science and Research, School of Pharmacy, Marshall University, One John Marshall Drive, Huntington, WV 25755, USA.
| | - Yunhui Xu
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA.
| | - Joel Turley
- Department of Pharmaceutical Science and Research, School of Pharmacy, Marshall University, One John Marshall Drive, Huntington, WV 25755, USA.
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA.
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA.
| | - Jinsong Hao
- Department of Pharmaceutical Science and Research, School of Pharmacy, Marshall University, One John Marshall Drive, Huntington, WV 25755, USA.
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA.
| |
Collapse
|
43
|
Kaur R, Sharma A, Sodhi M, Swami SK, Sharma VL, Kumari P, Verma P, Mukesh M. Sequence characterization of alpha 1 isoform (ATP1A1) of Na+/K+-ATPase gene and expression characteristics of its major isoforms across tissues of riverine buffaloes (Bubalus bubalis). GENE REPORTS 2018. [DOI: 10.1016/j.genrep.2017.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
44
|
Lopachev AV, Lopacheva OM, Nikiforova KA, Filimonov IS, Fedorova TN, Akkuratov EE. Comparative Action of Cardiotonic Steroids on Intracellular Processes in Rat Cortical Neurons. BIOCHEMISTRY (MOSCOW) 2018; 83:140-151. [DOI: 10.1134/s0006297918020062] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
45
|
Hodes A, Lifschytz T, Rosen H, Cohen Ben-Ami H, Lichtstein D. Reduction in endogenous cardiac steroids protects the brain from oxidative stress in a mouse model of mania induced by amphetamine. Brain Res Bull 2018; 137:356-362. [PMID: 29374602 DOI: 10.1016/j.brainresbull.2018.01.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/10/2018] [Accepted: 01/19/2018] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Bipolar disorder (BD) is a severe mental illness characterized by episodes of mania and depression. Numerous studies have implicated the involvement of endogenous cardiac steroids (CS), and their receptor, Na+, K+ -ATPase, in BD. The aim of the present study was to examine the role of brain oxidative stress in the CS-induced behavioral effects in mice. METHODS Amphetamine (AMPH)-induced hyperactivity, assessed in the open-field test, served as a model for manic-like behavior in mice. A reduction in brain CS was obtained by specific and sensitive anti-ouabain antibodies. The level of oxidative stress was tested in the hippocampus and frontal cortex by measuring the activity of antioxidant enzymes superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx), as well as the levels of antioxidant non-protein thiols (NPSH) and oxidative damage biomarkers thiobarbituric acid reactive substances (TBARS) and protein carbonyl (PC). RESULTS AMPH administration resulted in a marked hyperactivity and increased oxidative stress, as manifested by increased SOD activity, decreased activities of CAT and GPx, reduced levels of NPSH and increased levels of TBARS and PC. The administration of anti-ouabain antibodies, which reduced the AMPH-induced hyperactivity, protected against the concomitant oxidative stress in the brain. CONCLUSIONS Our results demonstrate that oxidative stress participates in the effects of endogenous CS on manic-like behavior induced by AMPH. These finding support the notion that CS and oxidative stress may be associated with the pathophysiology of mania and BD.
Collapse
Affiliation(s)
- Anastasia Hodes
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Tzuri Lifschytz
- Department of Psychiatry, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Haim Rosen
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Hagit Cohen Ben-Ami
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - David Lichtstein
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel.
| |
Collapse
|
46
|
Liu J, Lilly MN, Shapiro JI. Targeting Na/K-ATPase Signaling: A New Approach to Control Oxidative Stress. Curr Pharm Des 2018; 24:359-364. [PMID: 29318961 PMCID: PMC6052765 DOI: 10.2174/1381612824666180110101052] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 12/27/2017] [Accepted: 01/04/2017] [Indexed: 01/13/2023]
Abstract
Renal and cardiac function are greatly affected by chronic oxidative stress which can cause many pathophysiological states. The Na/K-ATPase is well-described as an ion pumping enzyme involved in maintaining cellular ion homeostasis; however, in the past two decades, extensive research has been done to understand the signaling function of the Na/K-ATPase and determine its role in physiological and pathophysiological states. Our lab has shown that the Na/K-ATPase signaling cascade can function as an amplifier of reactive oxygen species (ROS) which can be initiated by cardiotonic steroids or increases in ROS. Regulation of systemic oxidative stress by targeting Na/K-ATPase signaling mediated oxidant amplification improves 5/6th partial nephrectomy (PNx) mediated uremic cardiomyopathy, renal sodium handling, as well as ameliorates adipogenesis. This review will present this new concept of Na/K-ATPase signaling mediated oxidant amplification loop and its clinic implication.
Collapse
Affiliation(s)
- Jiang Liu
- Dept. of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | - Megan N. Lilly
- Dept. of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | - Joseph I. Shapiro
- Dept. of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| |
Collapse
|
47
|
Pereira DG, Salgado MA, Rocha SC, Santos HL, Villar JA, Contreras RG, Fontes CF, Barbosa LA, Cortes VF. Involvement of Src signaling in the synergistic effect between cisplatin and digoxin on cancer cell viability. J Cell Biochem 2017; 119:3352-3362. [DOI: 10.1002/jcb.26499] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 11/09/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Duane G. Pereira
- Faculdade de Bioquimica, Laboratorio de Bioquimica Celular, Universidade Federal de São João Del ReiCampus Centro‐Oeste Dona LinduDivinopolis, MGBrazil
| | - Mariana A.R. Salgado
- Faculdade de Bioquimica, Laboratorio de Bioquimica Celular, Universidade Federal de São João Del ReiCampus Centro‐Oeste Dona LinduDivinopolis, MGBrazil
| | - Sayonarah C. Rocha
- Faculdade de Bioquimica, Laboratorio de Bioquimica Celular, Universidade Federal de São João Del ReiCampus Centro‐Oeste Dona LinduDivinopolis, MGBrazil
| | - Hérica L. Santos
- Faculdade de Bioquimica, Laboratorio de Bioquimica Celular, Universidade Federal de São João Del ReiCampus Centro‐Oeste Dona LinduDivinopolis, MGBrazil
| | - José A.F.P. Villar
- Laboratório de Síntese Orgânica e Nanoestruturas, Universidade Federal de São João del ReiCampus Centro‐Oeste Dona LinduDivinopolis, MGBrazil
| | - Rubén G. Contreras
- Department of Physiology, Biophysics and NeurosciencesCenter for Research and Advanced Studies (Cinvestav)Mexico CityMexico
| | - Carlos F.L. Fontes
- Laboratório de Estrutura e Regulação de Proteínas e ATPases, Instituto de Bioquimica Médica Leopoldo de Meis, Centro de Ciências da SaúdeUniversidade Federal do Rio de JaneiroRio de Janeiro, RJBrazil
| | - Leandro A. Barbosa
- Faculdade de Bioquimica, Laboratorio de Bioquimica Celular, Universidade Federal de São João Del ReiCampus Centro‐Oeste Dona LinduDivinopolis, MGBrazil
| | - Vanessa F. Cortes
- Faculdade de Bioquimica, Laboratorio de Bioquimica Celular, Universidade Federal de São João Del ReiCampus Centro‐Oeste Dona LinduDivinopolis, MGBrazil
| |
Collapse
|
48
|
Lohr JN, Meinzer F, Dalla S, Romey-Glüsing R, Dobler S. The function and evolutionary significance of a triplicated Na,K-ATPase gene in a toxin-specialized insect. BMC Evol Biol 2017; 17:256. [PMID: 29246105 PMCID: PMC5732401 DOI: 10.1186/s12862-017-1097-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 11/27/2017] [Indexed: 01/27/2023] Open
Abstract
Background The Na,K-ATPase is a vital animal cell-membrane protein that maintains the cell’s resting potential, among other functions. Cardenolides, a group of potent plant toxins, bind to and inhibit this pump. The gene encoding the α-subunit of the pump has undergone duplication events in some insect species known to feed on plants containing cardenolides. Here we test the function of these duplicated gene copies in the cardenolide-adapted milkweed bug, Oncopeltus fasciatus, which has three known copies of the gene: α1A, α1B and α1C. Results Using RT-qPCR analyses we demonstrate that the α1C is highly expressed in neural tissue, where the pump is generally thought to be most important for neuron excitability. With the use of in vivo RNAi in adult bugs we found that α1C knockdowns suffered high mortality, where as α1A and α1B did not, supporting that α1C is most important for effective ion pumping. Next we show a role for α1A and α1B in the handling of cardenolides: expression results find that both copies are primarily expressed in the Malpighian tubules, the primary insect organ responsible for excretion, and when we injected either α1A or α1B knockdowns with cardenolides this proved fatal (whereas not in controls). Conclusions These results show that the Na,K-ATPα gene-copies have taken on diverse functions. Having multiple copies of this gene appears to have allowed the newly arisen duplicates to specialize on resistance to cardenolides, whereas the ancestral copy of the pump remains comparatively sensitive, but acts as a more efficient ion carrier. Interestingly both the α1A and α1B were required for cardenolide handling, suggesting that these two copies have separate and vital functions. Gene duplications of the Na,K-ATPase thus represent an excellent example of subfunctionalization in response to a new environmental challenge. Electronic supplementary material The online version of this article (10.1186/s12862-017-1097-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jennifer N Lohr
- Universität Hamburg, Biozentrum Grindel, Zoologisches Institut, Martin-Luther-King Pl. 3, 20146, Hamburg, Germany. .,Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, WC1E 6BT, London, UK.
| | - Fee Meinzer
- Universität Hamburg, Biozentrum Grindel, Zoologisches Institut, Martin-Luther-King Pl. 3, 20146, Hamburg, Germany
| | - Safaa Dalla
- Universität Hamburg, Biozentrum Grindel, Zoologisches Institut, Martin-Luther-King Pl. 3, 20146, Hamburg, Germany
| | - Renja Romey-Glüsing
- Universität Hamburg, Biozentrum Grindel, Zoologisches Institut, Martin-Luther-King Pl. 3, 20146, Hamburg, Germany
| | - Susanne Dobler
- Universität Hamburg, Biozentrum Grindel, Zoologisches Institut, Martin-Luther-King Pl. 3, 20146, Hamburg, Germany
| |
Collapse
|
49
|
Petrushanko IY, Mitkevich VA, Lakunina VA, Anashkina AA, Spirin PV, Rubtsov PM, Prassolov VS, Bogdanov NB, Hänggi P, Fuller W, Makarov AA, Bogdanova A. Cysteine residues 244 and 458-459 within the catalytic subunit of Na,K-ATPase control the enzyme's hydrolytic and signaling function under hypoxic conditions. Redox Biol 2017; 13:310-319. [PMID: 28601781 PMCID: PMC5470536 DOI: 10.1016/j.redox.2017.05.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 03/16/2017] [Accepted: 05/26/2017] [Indexed: 12/21/2022] Open
Abstract
Our previous findings suggested that reversible thiol modifications of cysteine residues within the actuator (AD) and nucleotide binding domain (NBD) of the Na,K-ATPase may represent a powerful regulatory mechanism conveying redox- and oxygen-sensitivity of this multifunctional enzyme. S-glutathionylation of Cys244 in the AD and Cys 454-458-459 in the NBD inhibited the enzyme and protected cysteines' thiol groups from irreversible oxidation under hypoxic conditions. In this study mutagenesis approach was used to assess the role these cysteines play in regulation of the Na,K-ATPase hydrolytic and signaling functions. Several constructs of mouse α1 subunit of the Na,K-ATPase were produced in which Cys244, Cys 454-458-459 or Cys 244-454-458-459 were replaced by alanine. These constructs were expressed in human HEK293 cells. Non-transfected cells and those expressing murine α1 subunit were exposed to hypoxia or treated with oxidized glutathione (GSSG). Both conditions induced inhibition of the wild type Na,K-ATPase. Enzymes containing mutated mouse α1 lacking Cys244 or all four cysteines (Cys 244-454-458-459) were insensitive to hypoxia. Inhibitory effect of GSSG was observed for wild type murine Na,K-ATPase, but was less pronounced in Cys454-458-459Ala mutant and completely absent in the Cys244Ala and Cys 244-454-458-459Ala mutants. In cells, expressing wild type enzyme, ouabain induced activation of Src and Erk kinases under normoxic conditions, whereas under hypoxic conditions this effect was inversed. Cys454-458-459Ala substitution abolished Src kinase activation in response to ouabain treatment, uncoupled Src from Erk signaling, and interfered with O2-sensitivity of Na,K-ATPase signaling function. Moreover, modeling predicted that S-glutathionylation of Cys 458 and 459 should prevent inhibitory binding of Src to NBD. Our data indicate for the first time that cysteine residues within the AD and NBD influence hydrolytic as well as receptor function of the Na,K-ATPase and alter responses of the enzyme to hypoxia or upon treatment with cardiotonic steroids.
Collapse
Affiliation(s)
- Irina Yu Petrushanko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Vladimir A Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Valentina A Lakunina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anastasia A Anashkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Pavel V Spirin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Peter M Rubtsov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Vladimir S Prassolov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Nikolay B Bogdanov
- Institute of Veterinary Physiology, Vetsuisse Faculty and the Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Pascal Hänggi
- Institute of Veterinary Physiology, Vetsuisse Faculty and the Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - William Fuller
- Cardiovascular and Diabetes Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anna Bogdanova
- Institute of Veterinary Physiology, Vetsuisse Faculty and the Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.
| |
Collapse
|
50
|
Rajamanickam GD, Kastelic JP, Thundathil JC. Na/K-ATPase regulates bovine sperm capacitation through raft- and non-raft-mediated signaling mechanisms. Mol Reprod Dev 2017; 84:1168-1182. [PMID: 28833817 DOI: 10.1002/mrd.22879] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 08/09/2017] [Indexed: 11/06/2022]
Abstract
Highly dynamic lipid microdomains (rafts) in the sperm plasma membrane contain several signaling proteins that regulate sperm capacitation. Na/K-ATPase isoforms (testis-specific isoform ATP1A4 and ubiquitous isoform ATP1A1) are abundant in bovine sperm plasma membrane. We previously reported that incubation of bovine sperm with ouabain, a specific Na/K-ATPase ligand, induced tyrosine phosphorylation of several sperm proteins during capacitation. The objective of this study was to investigate the roles of lipid rafts and non-rafts in Na/K-ATPase enzyme activity and signaling during bovine sperm capacitation. Content of ATP1A4 and, to a lesser extent, ATP1A1 was increased in raft and non-raft fractions of capacitated sperm, although non-raft enzyme activities of both isoforms were higher than the corresponding activities in rafts from capacitated sperm. Yet, ATP1A4 was the predominant isoform responsible for total Na/K-ATPase activity in both rafts and non-rafts. A comparative increase in phosphorylation of signaling molecules was observed in both raft (CAV1) and non-raft (EGFR and ERK1/2) membrane fractions during capacitation. Although SRC was phosphorylated in both membrane fractions, the non-raft fraction possessed more of this activated form. We also inferred, by immunoprecipitation, that ATP1A4 interacted with CAV1 and EGFR in the raft fraction, whereas interactions of ATP1A4 with SRC, EGFR, and ERK1/2 occurred in the non-raft fraction of ouabain-capacitated sperm; conversely, ATP1A1 interacted only with CAV1 in both fractions of uncapacitated and capacitated sperm. In conclusion, both raft and non-raft cohorts of Na/K-ATPase isoforms contributed to phosphorylation of signaling molecules during bovine sperm capacitation.
Collapse
Affiliation(s)
- Gayathri D Rajamanickam
- Faculty of Veterinary Medicine, Department of Production Animal Health, University of Calgary, Calgary, Alberta, Canada
| | - John P Kastelic
- Faculty of Veterinary Medicine, Department of Production Animal Health, University of Calgary, Calgary, Alberta, Canada
| | - Jacob C Thundathil
- Faculty of Veterinary Medicine, Department of Production Animal Health, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|