1
|
Orellana AM, Port's NMS, de Sá Lima L, Leite JA, Andreotti DZ, Kinoshita PF, Cantanzaro AB, Neto JAM, Scavone C, Kawamoto EM. Ouabain increases neuronal differentiation of hippocampal neural precursor cells. CURRENT RESEARCH IN NEUROBIOLOGY 2025; 8:100147. [PMID: 40166632 PMCID: PMC11957680 DOI: 10.1016/j.crneur.2025.100147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 04/02/2025] Open
Abstract
Image 1.
Collapse
Affiliation(s)
- Ana Maria Orellana
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Natacha Medeiros S. Port's
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Larissa de Sá Lima
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Jacqueline Alves Leite
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Goiás, 74045-155, Brazil
| | - Diana Zukas Andreotti
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Paula Fernanda Kinoshita
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Arthur B. Cantanzaro
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - João Agostinho M. Neto
- Laboratory of Cancer biology and Antineoplastic agents. Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Cristoforo Scavone
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Elisa M. Kawamoto
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| |
Collapse
|
2
|
Cordeiro GA, Faria JA, Pavan L, Garcia IJP, Neves EPFI, Lima GFDF, Campos HM, Ferreira PY, Ghedini PC, Kawamoto EM, Lima MC, Villar JAFP, Orellana AMM, Barbosa LA, Scavone C, Leite JA, Santos HL. Evaluation of the neuroprotective potential of benzylidene digoxin 15 against oxidative stress in a neuroinflammation models induced by lipopolysaccharide and on neuronal differentiation of hippocampal neural precursor cells. Front Pharmacol 2025; 16:1537720. [PMID: 40160463 PMCID: PMC11949953 DOI: 10.3389/fphar.2025.1537720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/12/2025] [Indexed: 04/02/2025] Open
Abstract
Neuroinflammation, often driven by the overproduction of reactive oxygen species (ROS), plays a crucial role in the pathogenesis of neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. The susceptibility of the brain to oxidative stress is attributed to its high metabolic activity and limited antioxidant defense. This study aimed to evaluate the neuroprotective potential of Benzylidene Digoxin 15 (BD-15) following treatment and pretreatment in a lipopolysaccharide (LPS)-induced neuroinflammation model. Additionally, we examined whether BD-15 enhances the generation of neurons from neural progenitor cells (NPCs).Male Wistar rats were used for acute treatment studies and divided into four groups: control (saline), BD-15 (100 μg/kg), LPS (250 μg/kg), and LPS + BD-15 (250 μg/kg + 100 μg/kg). Swiss albino mice were used for chronic pretreatment studies and divided into the following groups: control (saline), BD-15 (0.56 mg/kg), LPS (1 mg/kg), and LPS + BD-15 (1 mg/kg + 0.56 mg/kg). Behavioral changes were assessed using the open field test, and brain tissues were analyzed for oxidative stress markers, including malondialdehyde (MDA), reduced glutathione (GSH), protein carbonylation, catalase (CAT), superoxide dismutase (SOD), and glutathione S-transferase (GST). To assess neurogenesis, primary NPC cultures derived from the hippocampus of newborn Wistar rats were used, which led to reduced locomotor activity and increased oxidative stress, particularly in the cortex, as indicated by elevated MDA levels and reduced GSH levels. BD-15 treatment reversed these effects, notably by restoring GSH levels and reducing protein carbonylation in the cerebellum. Chronic BD-15 treatment in Swiss mice improved oxidative stress markers including MDA, SOD, CAT, and GST. Furthermore, BD-15 exhibits neuroprotective properties by alleviating oxidative stress and motor dysfunction, suggesting its potential as a therapeutic agent for neuroinflammatory disorders. However, BD-15 did not affect NPC cell proliferation, indicating that this cardiotonic steroid did not alter the cell cycle of these progenitor cells.
Collapse
Affiliation(s)
- Gilvânia A. Cordeiro
- Laboratório de Bioquímica Celular, UFSJ, Universidade Federal de São João del-Rei, Divinópolis, Minas Gerais, Brazil
| | - Jessica A. Faria
- Laboratório de Bioquímica Celular, UFSJ, Universidade Federal de São João del-Rei, Divinópolis, Minas Gerais, Brazil
| | | | - Israel J. P. Garcia
- Laboratório de Bioquímica Celular, UFSJ, Universidade Federal de São João del-Rei, Divinópolis, Minas Gerais, Brazil
| | - Eduarda P. F. I. Neves
- Instituto de Ciências Biológicas, UFG, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | | | - Hericles M. Campos
- Instituto de Ciências Biológicas, UFG, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Pâmela Y. Ferreira
- Instituto de Ciências Biológicas, UFG, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Paulo C. Ghedini
- Instituto de Ciências Biológicas, UFG, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | | | - Maira C. Lima
- Laboratório de Bioquímica Celular, UFSJ, Universidade Federal de São João del-Rei, Divinópolis, Minas Gerais, Brazil
| | - José A. F. P. Villar
- Laboratório de Bioquímica Celular, UFSJ, Universidade Federal de São João del-Rei, Divinópolis, Minas Gerais, Brazil
- Laboratório de Síntese Orgânica e Nanoestruturas, UFSJ, Universidade Federal de São João del-Rei, Divinópolis, Minas Gerais, Brazil
| | | | - Leandro A. Barbosa
- Laboratório de Bioquímica Celular, UFSJ, Universidade Federal de São João del-Rei, Divinópolis, Minas Gerais, Brazil
| | | | - Jacqueline A. Leite
- Instituto de Ciências Biológicas, UFG, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Hérica L. Santos
- Laboratório de Bioquímica Celular, UFSJ, Universidade Federal de São João del-Rei, Divinópolis, Minas Gerais, Brazil
| |
Collapse
|
3
|
Rasko NB, Nahm CB, Turchini J, Teh R, Rasmussen H, Byeon S, Sahni S, Samra JS, Gill AJ, Mittal A. FXYD3 Is Frequently Expressed in Pancreatic Ductal Adenocarcinoma but Does Not Predict Survival. Cancer Med 2025; 14:e70500. [PMID: 39783776 PMCID: PMC11714220 DOI: 10.1002/cam4.70500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 11/08/2024] [Accepted: 12/01/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND FXYD3 is a Na/K-ATPase modulator which is upregulated in pancreatic ductal adenocarcinoma (PDAC), but its prognostic role is unknown. This study evaluated FXYD3 expression in chemo-naive patients with surgically-resected PDAC at a single centre (1993-2014). METHOD FXYD3 expression was assessed in tumour specimens using immunohistochemistry. RESULTS 145 of 180 PDAC tumour specimens were FXYD3-immunopositive (80.5%). There was no difference in median overall survival between the FXYD3 negative (27.60 months) and positive groups (25.00 months) (log-rank p = 0.9718). FXYD3 expression correlated positively with late-stage disease (OR 3.041, 95% CI 1.190-7.455, p = 0.0175). There was no significant association with T stage, positive lymph nodes, perineural invasion, lymphovascular invasion or histological grade. CONCLUSION Immunohistochemical FXYD3 expression does not predict survival in chemo-naive PDAC patients, but is associated with late-stage disease. The high rate of FXYD3 overexpression warrants therapeutic evaluation.
Collapse
Affiliation(s)
- Nathalie B. Rasko
- Faculty of Medicine and Health, Sydney Medical SchoolUniversity of SydneySydneyNew South WalesAustralia
| | - Christopher B. Nahm
- Faculty of Medicine and Health, Sydney Medical SchoolUniversity of SydneySydneyNew South WalesAustralia
- Upper Gastrointestinal Surgical Unit, Department of Gastrointestinal SurgeryRoyal North Shore HospitalSydneyNew South WalesAustralia
| | - John Turchini
- Faculty of Medicine and Health, Sydney Medical SchoolUniversity of SydneySydneyNew South WalesAustralia
- Cancer Diagnosis and Pathology Group, Kolling InstituteRoyal North Shore HospitalSydneyNew South WalesAustralia
| | - Rachel Teh
- Faculty of Medicine and Health, Sydney Medical SchoolUniversity of SydneySydneyNew South WalesAustralia
| | - Helge Rasmussen
- Faculty of Medicine and Health, Sydney Medical SchoolUniversity of SydneySydneyNew South WalesAustralia
- Cardiac Membrane Biology Laboratory, Kolling InstituteThe University of SydneySydneyNew South WalesAustralia
| | - Sooin Byeon
- Faculty of Medicine and Health, Sydney Medical SchoolUniversity of SydneySydneyNew South WalesAustralia
| | - Sumit Sahni
- Faculty of Medicine and Health, Sydney Medical SchoolUniversity of SydneySydneyNew South WalesAustralia
- Australian Pancreatic CentreSydneyNew South WalesAustralia
| | - Jaswinder S. Samra
- Faculty of Medicine and Health, Sydney Medical SchoolUniversity of SydneySydneyNew South WalesAustralia
- Upper Gastrointestinal Surgical Unit, Department of Gastrointestinal SurgeryRoyal North Shore HospitalSydneyNew South WalesAustralia
- Australian Pancreatic CentreSydneyNew South WalesAustralia
- Faculty of Medical and Health SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Anthony J. Gill
- Faculty of Medicine and Health, Sydney Medical SchoolUniversity of SydneySydneyNew South WalesAustralia
- Cancer Diagnosis and Pathology Group, Kolling InstituteRoyal North Shore HospitalSydneyNew South WalesAustralia
| | - Anubhav Mittal
- Faculty of Medicine and Health, Sydney Medical SchoolUniversity of SydneySydneyNew South WalesAustralia
- Upper Gastrointestinal Surgical Unit, Department of Gastrointestinal SurgeryRoyal North Shore HospitalSydneyNew South WalesAustralia
- Australian Pancreatic CentreSydneyNew South WalesAustralia
| |
Collapse
|
4
|
Fujii F, Kanemasa H, Okuzono S, Setoyama D, Taira R, Yonemoto K, Motomura Y, Kato H, Masuda K, Kato TA, Ohga S, Sakai Y. ATP1A3 regulates protein synthesis for mitochondrial stability under heat stress. Dis Model Mech 2024; 17:dmm050574. [PMID: 38804677 PMCID: PMC11247502 DOI: 10.1242/dmm.050574] [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: 11/02/2023] [Accepted: 05/20/2024] [Indexed: 05/29/2024] Open
Abstract
Pathogenic variants in ATP1A3, the gene encoding the α3 subunit of the Na+/K+-ATPase, cause alternating hemiplegia of childhood (AHC) and related disorders. Impairments in Na+/K+-ATPase activity are associated with the clinical phenotype. However, it remains unclear whether additional mechanisms are involved in the exaggerated symptoms under stressed conditions in patients with AHC. We herein report that the intracellular loop (ICL) of ATP1A3 interacted with RNA-binding proteins, such as Eif4g (encoded by Eif4g1), Pabpc1 and Fmrp (encoded by Fmr1), in mouse Neuro2a cells. Both the siRNA-mediated depletion of Atp1a3 and ectopic expression of the p.R756C variant of human ATP1A3-ICL in Neuro2a cells resulted in excessive phosphorylation of ribosomal protein S6 (encoded by Rps6) and increased susceptibility to heat stress. In agreement with these findings, induced pluripotent stem cells (iPSCs) from a patient with the p.R756C variant were more vulnerable to heat stress than control iPSCs. Neurons established from the patient-derived iPSCs showed lower calcium influxes in responses to stimulation with ATP than those in control iPSCs. These data indicate that inefficient protein synthesis contributes to the progressive and deteriorating phenotypes in patients with the p.R756C variant among a variety of ATP1A3-related disorders.
Collapse
Affiliation(s)
- Fumihiko Fujii
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Hikaru Kanemasa
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Sayaka Okuzono
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Daiki Setoyama
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Ryoji Taira
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Kousuke Yonemoto
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yoshitomo Motomura
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Hiroki Kato
- Department of Molecular Cell Biology and Oral Anatomy, Graduate School of Dental Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Keiji Masuda
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Takahiro A. Kato
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yasunari Sakai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Jahanshahi S, Ouyang H, Ahmed C, Zahedi Amiri A, Dahal S, Mao YQ, Van Ommen DAJ, Malty R, Duan W, Been T, Hernandez J, Mangos M, Nurtanto J, Babu M, Attisano L, Houry WA, Moraes TJ, Cochrane A. Broad spectrum post-entry inhibitors of coronavirus replication: Cardiotonic steroids and monensin. Virology 2024; 589:109915. [PMID: 37931588 DOI: 10.1016/j.virol.2023.109915] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/10/2023] [Accepted: 10/17/2023] [Indexed: 11/08/2023]
Abstract
A small molecule screen identified several cardiotonic steroids (digitoxin and ouabain) and the ionophore monensin as potent inhibitors of HCoV-229E, HCoV-OC43, and SARS-CoV-2 replication with EC50s in the low nM range. Subsequent tests confirmed antiviral activity in primary cell models including human nasal epithelial cells and lung organoids. Addition of digitoxin, ouabain, or monensin strongly reduced viral gene expression as measured by both viral protein and RNA accumulation. Furthermore, the compounds acted post virus entry. While the antiviral activity of digitoxin was dependent upon activation of the MEK and JNK signaling pathways but not signaling through GPCRs, the antiviral effect of monensin was reversed upon inhibition of several signaling pathways. Together, the data demonstrates the potent anti-coronavirus properties of two classes of FDA approved drugs that function by altering the properties of the infected cell, rendering it unable to support virus replication.
Collapse
Affiliation(s)
- Shahrzad Jahanshahi
- Dept. of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada; Dept. of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Hong Ouyang
- Program in Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Choudhary Ahmed
- Dept. of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Ali Zahedi Amiri
- Dept. of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Subha Dahal
- Dept. of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Yu-Qian Mao
- Dept. of Biochemistry, University of Toronto, Toronto, ON, Canada
| | | | - Ramy Malty
- Dept. of Biochemistry, University of Toronto, Toronto, ON, Canada; Research and Innovation Centre, Department of Biochemistry, University of Regina, Regina, SK, Canada
| | - Wenming Duan
- Program in Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Terek Been
- Dept. of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | - Maria Mangos
- Donnelly Center, University of Toronto, Ontario, Canada
| | | | - Mohan Babu
- Research and Innovation Centre, Department of Biochemistry, University of Regina, Regina, SK, Canada
| | - Liliana Attisano
- Dept. of Biochemistry, University of Toronto, Toronto, ON, Canada; Donnelly Center, University of Toronto, Ontario, Canada
| | - Walid A Houry
- Dept. of Biochemistry, University of Toronto, Toronto, ON, Canada; Dept. of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Theo J Moraes
- Program in Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Alan Cochrane
- Dept. of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
8
|
Gurler B, Gencay G, Baloglu E. Hypoxia and HIF-1α Regulate the Activity and Expression of Na,K-ATPase Subunits in H9c2 Cardiomyoblasts. Curr Issues Mol Biol 2023; 45:8277-8288. [PMID: 37886965 PMCID: PMC10605391 DOI: 10.3390/cimb45100522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
The optimal function of the Na,K-ATPase (NKA) pump is essential for the heart. In ischemic heart disease, NKA activity decreases due to the decreased expression of the pump subunits. Here, we tested whether the hypoxia-inducible transcription factor (HIF-1α), the key signaling molecule regulating the adaptation of cells to hypoxia, is involved in controlling the expression and cellular dynamics of α1- and β1-NKA isoforms and of NKA activity in in-vitro hypoxic H9c2 cardiomyoblasts. HIF-1α was silenced through adenoviral infection, and cells were kept in normoxia (19% O2) or hypoxia (1% O2) for 24 h. We investigated the mRNA and protein expression of α1-, β1-NKA using RT-qPCR and Western blot in whole-cell lysates, cell membranes, and cytoplasmic fractions after labeling the cell surface with NHS-SS-biotin and immunoprecipitation. NKA activity and intracellular ATP levels were also measured. We found that in hypoxia, silencing HIF-1α prevented the decreased mRNA expression of α1-NKA but not of β1-NKA. Hypoxia decreased the plasma membrane expression of α1-NKA and β1- NKA compared to normoxic cells. In hypoxic cells, HIF-1α silencing prevented this effect by inhibiting the internalization of α1-NKA. Total protein expression was not affected. The decreased activity of NKA in hypoxic cells was fully prevented by silencing HIF-1α independent of cellular ATP levels. This study is the first to show that in hypoxic H9c2 cardiomyoblasts, HIF-1α controls the internalization and membrane insertion of α1-NKA subunit and of NKA activity. The mechanism behind this regulation needs further investigation.
Collapse
Affiliation(s)
- Beyza Gurler
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey;
| | - Gizem Gencay
- Department of Molecular and Translational Biomedicine, Institute of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey;
| | - Emel Baloglu
- Department of Medical Pharmacology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| |
Collapse
|
9
|
Tomazelli CA, Ishikawa FM, Couto GK, Parente JM, Castro MMD, Xavier FE, Rossoni LV. Small artery remodeling and stiffening in deoxycorticosterone acetate-salt hypertensive rats involves the interaction between endogenous ouabain/Na + K + -ATPase/cSrc signaling. J Hypertens 2023; 41:1554-1564. [PMID: 37432904 DOI: 10.1097/hjh.0000000000003502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
OBJECTIVE Endogenous ouabain (EO) increases in some patients with hypertension and in rats with volume-dependent hypertension. When ouabain binds to Na + K + -ATPase, cSrc is activated, which leads to multieffector signaling activation and high blood pressure (BP). In mesenteric resistance arteries (MRA) from deoxycorticosterone acetate (DOCA)-salt rats, we have demonstrated that the EO antagonist rostafuroxin blocks downstream cSrc activation, enhancing endothelial function and lowering oxidative stress and BP. Here, we examined the possibility that EO is involved in the structural and mechanical alterations that occur in MRA from DOCA-salt rats. METHODS MRA were taken from control, vehicle-treated DOCA-salt or rostafuroxin (1 mg/kg per day, for 3 weeks)-treated DOCA-salt rats. Pressure myography and histology were used to evaluate the mechanics and structure of the MRA, and western blotting to assess protein expression. RESULTS DOCA-salt MRA exhibited signs of inward hypertrophic remodeling and increased stiffness, with a higher wall:lumen ratio, which were reduced by rostafuroxin treatment. The enhanced type I collagen, TGFβ1, pSmad2/3 Ser465/457 /Smad2/3 ratio, CTGF, p-Src Tyr418 , EGFR, c-Raf, ERK1/2 and p38MAPK protein expression in DOCA-salt MRA were all recovered by rostafuroxin. CONCLUSION A process combining Na + K + -ATPase/cSrc/EGFR/Raf/ERK1/2/p38MAPK activation and a Na + K + -ATPase/cSrc/TGF-1/Smad2/3/CTGF-dependent mechanism explains how EO contributes to small artery inward hypertrophic remodeling and stiffening in DOCA-salt rats. This result supports the significance of EO as a key mediator for end-organ damage in volume-dependent hypertension and the efficacy of rostafuroxin in avoiding remodeling and stiffening of small arteries.
Collapse
Affiliation(s)
| | | | | | | | | | - Fabiano Elias Xavier
- Department of Physiology and Pharmacology, Biosciences Center, Federal University of Pernambuco, Recife, Brazil
| | | |
Collapse
|
10
|
Carullo N, Fabiano G, D'Agostino M, Zicarelli MT, Musolino M, Presta P, Michael A, Andreucci M, Bolignano D, Coppolino G. New Insights on the Role of Marinobufagenin from Bench to Bedside in Cardiovascular and Kidney Diseases. Int J Mol Sci 2023; 24:11186. [PMID: 37446363 DOI: 10.3390/ijms241311186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Marinobufagenin (MBG) is a member of the bufadienolide family of compounds, which are natural cardiac glycosides found in a variety of animal species, including man, which have different physiological and biochemical functions but have a common action on the inhibition of the adenosine triphosphatase sodium-potassium pump (Na+/K+-ATPase). MBG acts as an endogenous cardiotonic steroid, and in the last decade, its role as a pathogenic factor in various human diseases has emerged. In this paper, we have collated major evidence regarding the biological characteristics and functions of MBG and its implications in human pathology. This review focused on MBG involvement in chronic kidney disease, including end-stage renal disease, cardiovascular diseases, sex and gender medicine, and its actions on the nervous and immune systems. The role of MBG in pathogenesis and the development of a wide range of pathological conditions indicate that this endogenous peptide could be used in the future as a diagnostic biomarker and/or therapeutic target, opening important avenues of scientific research.
Collapse
Affiliation(s)
- Nazareno Carullo
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Giuseppe Fabiano
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Mario D'Agostino
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | | | - Michela Musolino
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Pierangela Presta
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Ashour Michael
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Michele Andreucci
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Davide Bolignano
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Giuseppe Coppolino
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
11
|
Rogachevskii IV, Samosvat DM, Penniyaynen VA, Plakhova VB, Podzorova SA, Ma K, Zegrya GG, Krylov BV. Role of the Rhamnosyl Residue of Ouabain in the Activation of the Na,K-ATPase Signaling Function. Life (Basel) 2023; 13:1500. [PMID: 37511875 PMCID: PMC10381505 DOI: 10.3390/life13071500] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/13/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
The signaling or non-pumping Na,K-ATPase function was first observed by us in the nociceptive neuron; Na,K-ATPase transduced the signals from the opioid-like receptors to NaV1.8 channels. This study elucidates the role of the rhamnosyl residue of ouabain in the activation of the Na,K-ATPase signaling function. The effects resulting from activation of Na,K-ATPase signaling by the Ca2+ chelate complex of ouabain (EO) are not manifested upon removal of the rhamnosyl residue, as demonstrated in viable cells by the highly sensitive patch-clamp and organotypic cell culture methods. Docking calculations show that the rhamnosyl residue is involved in five intermolecular hydrogen bonds with the Na,K-ATPase α1-subunit, which are fundamentally important for activation of the Na,K-ATPase signaling function upon EO binding. The main contribution to the energy of EO binding is provided by its steroid core, which forms a number of hydrogen bonds and hydrophobic interactions with Na,K-ATPase that stabilize the ligand-receptor complex. Another critically important role in EO binding is expected to be played by the chelated Ca2+ cation, which should switch on strong intermolecular ionic interactions between the EO molecule and two α1-Na,K-ATPase amino acid residues, Glu116 and Glu117.
Collapse
Affiliation(s)
- Ilya V Rogachevskii
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 199034 Saint Petersburg, Russia
| | - Dmitriy M Samosvat
- Ioffe Institute of the Russian Academy of Sciences, 194021 Saint Petersburg, Russia
| | - Valentina A Penniyaynen
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 199034 Saint Petersburg, Russia
| | - Vera B Plakhova
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 199034 Saint Petersburg, Russia
| | - Svetlana A Podzorova
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 199034 Saint Petersburg, Russia
| | - Ke Ma
- Department of Pain Management, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200240, China
| | - Georgy G Zegrya
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 199034 Saint Petersburg, Russia
- Ioffe Institute of the Russian Academy of Sciences, 194021 Saint Petersburg, Russia
| | - Boris V Krylov
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 199034 Saint Petersburg, Russia
| |
Collapse
|
12
|
Wu KX, Yogarajah T, Choy Loe MW, Kaur P, Hua Lee RC, Mok CK, Wong YH, Phuektes P, Yeo LS, Chow VT, Tan YW, Hann Chu JJ. The host-targeting compound peruvoside has a broad-spectrum antiviral activity against positive-sense RNA viruses. Acta Pharm Sin B 2023; 13:2039-2055. [DOI: 10.1016/j.apsb.2023.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/22/2023] [Accepted: 03/02/2023] [Indexed: 03/19/2023] Open
|
13
|
Gokula V, Terrero D, Joe B. Six Decades of History of Hypertension Research at the University of Toledo: Highlighting Pioneering Contributions in Biochemistry, Genetics, and Host-Microbiota Interactions. Curr Hypertens Rep 2022; 24:669-685. [PMID: 36301488 PMCID: PMC9708772 DOI: 10.1007/s11906-022-01226-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW The study aims to capture the history and lineage of hypertension researchers from the University of Toledo in Ohio and showcase their collective scientific contributions dating from their initial discoveries of the physiology of adrenal and renal systems and genetics regulating blood pressure (BP) to its more contemporary contributions including microbiota and metabolomic links to BP regulation. RECENT FINDINGS The University of Toledo College of Medicine and Life Sciences (UTCOMLS), previously known as the Medical College of Ohio, has contributed significantly to our understanding of the etiology of hypertension. Two of the scientists, Patrick Mulrow and John Rapp from UTCOMLS, have been recognized with the highest honor, the Excellence in Hypertension award from the American Heart Association for their pioneering work on the physiology and genetics of hypertension, respectively. More recently, Bina Joe has continued their legacy in the basic sciences by uncovering previously unknown novel links between microbiota and metabolites to the etiology of hypertension, work that has been recognized by the American Heart Association with multiple awards. On the clinical research front, Christopher Cooper and colleagues lead the CORAL trials and contributed importantly to the investigations on renal artery stenosis treatment paradigms. Hypertension research at this institution has not only provided these pioneering insights, but also grown careers of scientists as leaders in academia as University Presidents and Deans of Medical Schools. Through the last decade, the university has expanded its commitment to Hypertension research as evident through the development of the Center for Hypertension and Precision Medicine led by Bina Joe as its founding Director. Hypertension being the top risk factor for cardiovascular diseases, which is the leading cause of human mortality, is an important area of research in multiple international universities. The UTCOMLS is one such university which, for the last 6 decades, has made significant contributions to our current understanding of hypertension. This review is a synthesis of this rich history. Additionally, it also serves as a collection of audio archives by more recent faculty who are also prominent leaders in the field of hypertension research, including John Rapp, Bina Joe, and Christopher Cooper, which are cataloged at Interviews .
Collapse
Affiliation(s)
- Veda Gokula
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo College of Medicine and Life Sciences, Block Health Science Building, 3000 Arlington Ave, Toledo, OH, 43614-2598, USA
| | - David Terrero
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy, University of Toledo, Toledo, OH, USA
| | - Bina Joe
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo College of Medicine and Life Sciences, Block Health Science Building, 3000 Arlington Ave, Toledo, OH, 43614-2598, USA.
| |
Collapse
|
14
|
Chemistry and the Potential Antiviral, Anticancer, and Anti-Inflammatory Activities of Cardiotonic Steroids Derived from Toads. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196586. [PMID: 36235123 PMCID: PMC9571018 DOI: 10.3390/molecules27196586] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/30/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022]
Abstract
Cardiotonic steroids (CTS) were first documented by ancient Egyptians more than 3000 years ago. Cardiotonic steroids are a group of steroid hormones that circulate in the blood of amphibians and toads and can also be extracted from natural products such as plants, herbs, and marines. It is well known that cardiotonic steroids reveal effects against congestive heart failure and atrial fibrillation; therefore, the term "cardiotonic" has been coined. Cardiotonic steroids are divided into two distinct groups: cardenolides (plant-derived) and bufadienolides (mainly of animal origin). Cardenolides have an unsaturated five-membered lactone ring attached to the steroid nucleus at position 17; bufadienolides have a doubly unsaturated six-membered lactone ring. Cancer is a leading cause of mortality in humans all over the world. In 2040, the global cancer load is expected to be 28.4 million cases, which would be a 47% increase from 2020. Moreover, viruses and inflammations also have a very nebative impact on human health and lead to mortality. In the current review, we focus on the chemistry, antiviral and anti-cancer activities of cardiotonic steroids from the naturally derived (toads) venom to combat these chronic devastating health problems. The databases of different research engines (Google Scholar, PubMed, Science Direct, and Sci-Finder) were screened using different combinations of the following terms: “cardiotonic steroids”, “anti-inflammatory”, “antiviral”, “anticancer”, “toad venom”, “bufadienolides”, and “poison chemical composition”. Various cardiotonic steroids were isolated from diverse toad species and exhibited superior anti-inflammatory, anticancer, and antiviral activities in in vivo and in vitro models such as marinobufagenin, gammabufotalin, resibufogenin, and bufalin. These steroids are especially difficult to identify. However, several compounds and their bioactivities were identified by using different molecular and biotechnological techniques. Biotechnology is a new tool to fully or partially generate upscaled quantities of natural products, which are otherwise only available at trace amounts in organisms.
Collapse
|
15
|
Tiwari S, Rajamanickam G, Unnikrishnan V, Ojaghi M, Kastelic JP, Thundathil JC. Testis-Specific Isoform of Na +-K + ATPase and Regulation of Bull Fertility. Int J Mol Sci 2022; 23:7936. [PMID: 35887284 PMCID: PMC9317330 DOI: 10.3390/ijms23147936] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 12/10/2022] Open
Abstract
An advanced understanding of sperm function is relevant for evidence-based male fertility prediction and addressing male infertility. A standard breeding soundness evaluation (BSE) merely identifies gross abnormalities in bulls, whereas selection based on single nucleotide polymorphisms and genomic estimated breeding values overlooks sub-microscopic differences in sperm. Molecular tools are important for validating genomic selection and advancing knowledge on the regulation of male fertility at an interdisciplinary level. Therefore, research in this field is now focused on developing a combination of in vitro sperm function tests and identifying biomarkers such as sperm proteins with critical roles in fertility. The Na+-K+ ATPase is a ubiquitous transmembrane protein and its α4 isoform (ATP1A4) is exclusively expressed in germ cells and sperm. Furthermore, ATP1A4 is essential for male fertility, as it interacts with signaling molecules in both raft and non-raft fractions of the sperm plasma membrane to regulate capacitation-associated signaling, hyperactivation, sperm-oocyte interactions, and activation. Interestingly, ATP1A4 activity and expression increase during capacitation, challenging the widely accepted dogma of sperm translational quiescence. This review discusses the literature on the role of ATP1A4 during capacitation and fertilization events and its prospective use in improving male fertility prediction.
Collapse
Affiliation(s)
| | | | | | | | | | - Jacob C. Thundathil
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (S.T.); (G.R.); (V.U.); (M.O.); (J.P.K.)
| |
Collapse
|
16
|
Bolignano D, De Rosa S, Greco M, Presta P, Patella G, Crugliano G, Sabatino J, Strangio A, Romano LR, Comi A, Cianfrone P, Andreucci M, Dragone F, Indolfi C, Foti DP, Coppolino G. Marinobufagenin, left ventricular geometry and cardiac dysfunction in end-stage kidney disease patients. Int Urol Nephrol 2022; 54:2581-2589. [PMID: 35274285 DOI: 10.1007/s11255-022-03161-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 02/23/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Left ventricular hypertrophy (LVH) is remarkably prevalent among end-stage kidney disease (ESKD) on chronic dialysis and has a strong prognostic value for adverse outcomes. In experimental models, the endogenous cardiotonic steroid Marinobufagenin (MBG) promotes cardiac hypertrophy and accelerates uremic cardiomyopathy. In this study, we investigated the possible relationships between MBG, LV geometry and cardiac dysfunction in a clinical setting of ESKD. METHODS Plasmatic MBG was measured in 46 prevalent ESKD patients (n = 30 HD, n = 16 PD) together with a thorough laboratory, clinical, bioimpedance and echocardiography assessment. Different patterns of LV geometry were defined by left ventricular mass index (LVMi) and ventricular morphology. Diastolic dysfunction was diagnosed by the ASE/EACVI criteria. RESULTS MBG levels were significantly higher in ESKD patients than in healthy controls (p = 0.001) and more elevated in PD than in HD (p = 0.02). At multivariate analyses, E/e' (β = 0.38; p = 0.009) and LVMi (β = 0.42; p = 0.02) remained the sole independent predictors of MBG. A statistically significant trend in MBG levels (p = 0.01) was noticed across different patterns of LV geometry, with the highest values found in eccentric LVH. MBG levels were higher in the presence of diastolic dysfunction (p = 0.01) and this substance displayed a remarkable diagnostic capacity in distinguish patients with normal LV geometry, LV hypertrophy and, particularly, eccentric LVH (AUC 0.888; p < 0.0001) and diastolic dysfunction (AUC 0.79; p = 0.001). CONCLUSIONS Deranged plasma MBG levels in ESKD patients on chronic dialysis reflect alterations in LV structure and function. MBG may, thus, candidate as a novel biomarker for improving cardiac assessment in this high-risk population.
Collapse
Affiliation(s)
- Davide Bolignano
- Nephrology and Dialysis Unit, Magna Graecia University of Catanzaro, Catanzaro, Italy. .,Department of Medical and Surgical Sciences-Renal Unit, Magna Graecia University of Catanzaro, Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy.
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences-Renal Unit, Magna Graecia University of Catanzaro, Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy.,Cardiovascular Research Center, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Marta Greco
- Clinical Pathology Lab, Magna Graecia University of Catanzaro, Catanzaro, Italy.,Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Pierangela Presta
- Nephrology and Dialysis Unit, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Gemma Patella
- Nephrology and Dialysis Unit, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Giuseppina Crugliano
- Nephrology and Dialysis Unit, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Jolanda Sabatino
- Department of Medical and Surgical Sciences-Renal Unit, Magna Graecia University of Catanzaro, Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy.,Cardiovascular Research Center, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Antonio Strangio
- Department of Medical and Surgical Sciences-Renal Unit, Magna Graecia University of Catanzaro, Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Letizia Rosa Romano
- Department of Medical and Surgical Sciences-Renal Unit, Magna Graecia University of Catanzaro, Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Alessandro Comi
- Nephrology and Dialysis Unit, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Paola Cianfrone
- Nephrology and Dialysis Unit, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Michele Andreucci
- Nephrology and Dialysis Unit, Magna Graecia University of Catanzaro, Catanzaro, Italy.,Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Francesco Dragone
- Clinical Pathology Lab, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Ciro Indolfi
- Department of Medical and Surgical Sciences-Renal Unit, Magna Graecia University of Catanzaro, Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy.,Cardiovascular Research Center, Magna Graecia University of Catanzaro, Catanzaro, Italy.,Mediterranea Cardiocentro, Naples, Italy
| | - Daniela Patrizia Foti
- Clinical Pathology Lab, Magna Graecia University of Catanzaro, Catanzaro, Italy.,Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Giuseppe Coppolino
- Nephrology and Dialysis Unit, Magna Graecia University of Catanzaro, Catanzaro, Italy.,Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy
| |
Collapse
|
17
|
Shandell MA, Capatina AL, Lawrence SM, Brackenbury WJ, Lagos D. Inhibition of the Na +/K +-ATPase by cardiac glycosides suppresses expression of the IDO1 immune checkpoint in cancer cells by reducing STAT1 activation. J Biol Chem 2022; 298:101707. [PMID: 35150740 PMCID: PMC8902613 DOI: 10.1016/j.jbc.2022.101707] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/18/2022] Open
Abstract
Despite extensive basic and clinical research on immune checkpoint regulatory pathways, little is known about the effects of the ionic tumor microenvironment on immune checkpoint expression and function. Here we describe a mechanistic link between Na+/K+-ATPase (NKA) inhibition and activity of the immune checkpoint protein indoleamine-pyrrole 2',3'-dioxygenase 1 (IDO1). We found that IDO1 was necessary and sufficient for production of kynurenine, a downstream tryptophan metabolite, in cancer cells. We developed a spectrophotometric assay to screen a library of 31 model ion transport-targeting compounds for potential effects on IDO1 function in A549 lung and MDA-MB-231 breast cancer cells. This revealed that the cardiac glycosides ouabain and digoxin inhibited kynurenine production at concentrations that did not affect cell survival. NKA inhibition by ouabain and digoxin resulted in increased intracellular Na+ levels and downregulation of IDO1 mRNA and protein levels, which was consistent with the reduction in kynurenine levels. Knockdown of ATP1A1, the ɑ1 subunit of the NKA and target of cardiac glycosides, increased Na+ levels to a lesser extent than cardiac glycoside treatment and did not affect IDO1 expression. However, ATP1A1 knockdown significantly enhanced the effect of cardiac glycosides on IDO1 expression and kynurenine production. Mechanistically, we show that cardiac glycoside treatment resulted in curtailing the length of phosphorylation-mediated stabilization of STAT1, a transcriptional regulator of IDO1 expression, an effect enhanced by ATP1A1 knockdown. Our findings reveal cross talk between ionic modulation via cardiac glycosides and immune checkpoint protein expression in cancer cells with broad mechanistic and clinical implications.
Collapse
Affiliation(s)
- Mia A Shandell
- Department of Biology, University of York, York, United Kingdom; Hull York Medical School, University of York, York, United Kingdom; York Biomedical Research Institute, University of York, York, United Kingdom
| | - Alina L Capatina
- Department of Biology, University of York, York, United Kingdom; York Biomedical Research Institute, University of York, York, United Kingdom
| | | | - William J Brackenbury
- Department of Biology, University of York, York, United Kingdom; York Biomedical Research Institute, University of York, York, United Kingdom
| | - Dimitris Lagos
- Hull York Medical School, University of York, York, United Kingdom; York Biomedical Research Institute, University of York, York, United Kingdom.
| |
Collapse
|
18
|
Role of Na +/K +-ATPase in ischemic stroke: in-depth perspectives from physiology to pharmacology. J Mol Med (Berl) 2021; 100:395-410. [PMID: 34839371 DOI: 10.1007/s00109-021-02143-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 08/27/2021] [Accepted: 09/20/2021] [Indexed: 12/26/2022]
Abstract
Na+/K+-ATPase (NKA) is a large transmembrane protein expressed in all cells. It is well studied for its ion exchanging function, which is indispensable for the maintenance of electrochemical gradients across the plasma membrane and herein neuronal excitability. The widely recognized pump function of NKA closely depends on its unique structure features and conformational changes upon binding of specific ions. Various Na+-dependent secondary transport systems are rigorously controlled by the ionic gradients generated by NKA and are essential for multiple physiological processes. In addition, roles of NKA as a signal transducer have also been unveiled nowadays. Plethora of signaling cascades are defined including Src-Ras-MAPK signaling, IP3R-mediated calcium oscillation, inflammation, and autophagy though most underlying mechanisms remain elusive. Ischemic stroke occurs when the blood flow carrying nutrients and oxygen into the brain is disrupted by blood clots, which is manifested by excitotoxicity, oxidative stress, inflammation, etc. The protective effect of NKA against ischemic stress is emerging gradually with the application of specific NKA inhibitor. However, NKA-related research is limited due to the opposite effects caused by NKA inhibitor at lower doses. The present review focuses on the recent progression involving different aspects about NKA in cellular homeostasis to present an in-depth understanding of this unique protein. Moreover, essential roles of NKA in ischemic pathology are discussed to provide a platform and bright future for the improvement in clinical research on ischemic stroke.
Collapse
|
19
|
Quo vadis Cardiac Glycoside Research? Toxins (Basel) 2021; 13:toxins13050344. [PMID: 34064873 PMCID: PMC8151307 DOI: 10.3390/toxins13050344] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 12/16/2022] Open
Abstract
Cardiac glycosides (CGs), toxins well-known for numerous human and cattle poisoning, are natural compounds, the biosynthesis of which occurs in various plants and animals as a self-protective mechanism to prevent grazing and predation. Interestingly, some insect species can take advantage of the CG’s toxicity and by absorbing them, they are also protected from predation. The mechanism of action of CG’s toxicity is inhibition of Na+/K+-ATPase (the sodium-potassium pump, NKA), which disrupts the ionic homeostasis leading to elevated Ca2+ concentration resulting in cell death. Thus, NKA serves as a molecular target for CGs (although it is not the only one) and even though CGs are toxic for humans and some animals, they can also be used as remedies for various diseases, such as cardiovascular ones, and possibly cancer. Although the anticancer mechanism of CGs has not been fully elucidated, yet, it is thought to be connected with the second role of NKA being a receptor that can induce several cell signaling cascades and even serve as a growth factor and, thus, inhibit cancer cell proliferation at low nontoxic concentrations. These growth inhibitory effects are often observed only in cancer cells, thereby, offering a possibility for CGs to be repositioned for cancer treatment serving not only as chemotherapeutic agents but also as immunogenic cell death triggers. Therefore, here, we report on CG’s chemical structures, production optimization, and biological activity with possible use in cancer therapy, as well as, discuss their antiviral potential which was discovered quite recently. Special attention has been devoted to digitoxin, digoxin, and ouabain.
Collapse
|
20
|
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 PMCID: PMC7919863 DOI: 10.1124/molpharm.120.000101] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [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
|
21
|
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
|
22
|
Plakhova VB, Penniyaynen VA, Rogachevskii IV, Podzorova SA, Khalisov MM, Ankudinov AV, Krylov BV. Dual mechanism of modulation of Na V1.8 sodium channels by ouabain. Can J Physiol Pharmacol 2020; 98:785-802. [PMID: 32687732 DOI: 10.1139/cjpp-2020-0197] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the primary sensory neuron, ouabain activates the dual mechanism that modulates the functional activity of NaV1.8 channels. Ouabain at endogenous concentrations (EO) triggers two different signaling cascades, in which the Na,K-ATPase/Src complex is the EO target and the signal transducer. The fast EO effect is based on modulation of the NaV1.8 channel activation gating device. EO triggers the tangential signaling cascade along the neuron membrane from Na,K-ATPase to the NaV1.8 channel. It evokes a decrease in effective charge transfer of the NaV1.8 channel activation gating device. Intracellular application of PP2, an inhibitor of Src kinase, completely eliminated the effect of EO, thus indicating the absence of direct EO binding to the NaV1.8 channel. The delayed EO effect probably controls the density of NaV1.8 channels in the neuron membrane. EO triggers the downstream signaling cascade to the neuron genome, which should result in a delayed decrease in the NaV1.8 channels' density. PKC and p38 MAPK are involved in this pathway. Identification of the dual mechanism of the strong EO effect on NaV1.8 channels makes it possible to suggest that application of EO to the primary sensory neuron membrane should result in a potent antinociceptive effect at the organismal level.
Collapse
Affiliation(s)
- Vera B Plakhova
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., 199034, Saint Petersburg, Russia
| | - Valentina A Penniyaynen
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., 199034, Saint Petersburg, Russia
| | - Ilia V Rogachevskii
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., 199034, Saint Petersburg, Russia
| | - Svetlana A Podzorova
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., 199034, Saint Petersburg, Russia
| | - Maksim M Khalisov
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., 199034, Saint Petersburg, Russia
| | - Alexander V Ankudinov
- Ioffe Physical Technical Institute, Russian Academy of Sciences, 26 Polytekhnicheskaya str., 194021, Saint Petersburg, Russia
| | - Boris V Krylov
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., 199034, Saint Petersburg, Russia
| |
Collapse
|
23
|
Aperia A, Brismar H, Uhlén P. Mending Fences: Na,K-ATPase signaling via Ca 2+ in the maintenance of epithelium integrity. Cell Calcium 2020; 88:102210. [PMID: 32380435 DOI: 10.1016/j.ceca.2020.102210] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/18/2022]
Abstract
Na,K-ATPase is a ubiquitous multifunctional protein that acts both as an ion pump and as a signal transducer. The signaling function is activated by ouabain in non-toxic concentrations. In epithelial cells the ouabain-bound Na,K-ATPase connects with the inositol 1,4,5-trisphosphate receptor via a short linear motif to activate low frequency Ca2+ oscillations. Within a couple of minutes this ouabain mediated signal has resulted in phosphorylation or dephosphorylation of 2580 phospho-sites. Proteins that control cell proliferation and cell adhesion and calmodulin regulated proteins are enriched among the ouabain phosphor-regulated proteins. The inositol 1,4,5-trisphosphate receptor and the stromal interaction molecule, which are both essential for the initiation of Ca2+ oscillations, belong to the ouabain phosphor-regulated proteins. Downstream effects of the ouabain-evoked Ca2+ signal in epithelial cells include interference with the intrinsic mitochondrial apoptotic process and stimulation of embryonic growth processes. The dual function of Na,K-ATPase as an ion pump and a signal transducer is now well established and evaluation of the physiological and pathophysiological consequences of this universal signal emerges as an urgent topic for future studies.
Collapse
Affiliation(s)
- Anita Aperia
- Science for Life Laboratory, Dept of Women's and Children's Health, Karolinska Institutet, Sweden
| | - Hjalmar Brismar
- Science for Life Laboratory, Dept of Women's and Children's Health, Karolinska Institutet, Sweden; Science for Life Laboratory, Dept of Applied Physics, KTH Royal Institute of Technology, Sweden.
| | - Per Uhlén
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Sweden
| |
Collapse
|
24
|
Cardiac glycosides with target at direct and indirect interactions with nuclear receptors. Biomed Pharmacother 2020; 127:110106. [PMID: 32248001 DOI: 10.1016/j.biopha.2020.110106] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 12/15/2022] Open
Abstract
Cardiac glycosides are compounds isolated from plants and animals and have been known since ancient times. These compounds inhibit the activity of the sodium potassium pump in eukaryotic cells. Cardiac glycosides were used as drugs in heart ailments to increase myocardial contraction force and, at the same time, to lower frequency of this contraction. An increasing number of studies have indicated that the biological effects of these compounds are not limited to inhibition of sodium-potassium pump activity. Furthermore, an increasing number of data have shown that they are synthesized in tissues of mammals, where they may act as a new class of steroid hormones or other hormones by mimicry to modulate various signaling pathways and influence whole organisms. Thus, we discuss the interactions of cardiac glycosides with the nuclear receptor superfamily of transcription factors activated by low-weight molecular ligands (including hormones) that regulate many functions of cells and organisms. Cardiac glycosides of endogenous and exogenous origin by interacting with nuclear receptors can affect the processes regulated by these transcription factors, including hormonal management, immune system, body defense, and carcinogenesis. They can also be treated as initial structures for combinatorial chemistry to produce new compounds (including drugs) with the desired properties.
Collapse
|
25
|
Pessôa MTC, Valadares JMM, Rocha SC, Silva SC, McDermott JP, Sánchez G, Varotti FP, Scavone C, Ribeiro RIMA, Villar JAFP, Blanco G, Barbosa LA. 21-Benzylidene digoxin decreases proliferation by inhibiting the EGFR/ERK signaling pathway and induces apoptosis in HeLa cells. Steroids 2020; 155:108551. [PMID: 31812624 PMCID: PMC7028499 DOI: 10.1016/j.steroids.2019.108551] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/30/2019] [Accepted: 12/02/2019] [Indexed: 12/12/2022]
Abstract
Cardiotonic steroids (CTS) are agents traditionally known for their capacity to bind to the Na,K-ATPase (NKA), affecting the ion transport and the contraction of the heart. Natural CTS have been shown to also have effects on cell signaling pathways. With the goal of developing a new CTS derivative, we synthesized a new digoxin derivative, 21-benzylidene digoxin (21-BD). Previously, we have shown that this compound binds to NKA and has cytotoxic actions on cancer, but not on normal cells. Here, we further studied the mechanisms of actions of 21-BD. Working with HeLa cells, we found that 21-BD decreases the basal, as well as the insulin stimulated proliferation. 21-BD reduces phosphorylation of the epidermal growth factor receptor (EGFR) and extracellular-regulated kinase (ERK), which are involved in pathways that stimulate cell proliferation. In addition, 21-BD promotes apoptosis, which is mediated by the translocation of Bax from the cytosol to mitochondria and the release of mitochondrial cytochrome c to the cytosol. 21-BD also activated caspases-8, -9 and -3, and induced the cleavage of poly (ADP-ribose) polymerase-1 (PARP-1). Altogether, these results show that the new compound that we have synthesized exerts cytotoxic actions on HeLa cells by inhibition of cell proliferation and the activation of both the extrinsic and intrinsic apoptotic pathways. These results support the relevance of the cardiotonic steroid scaffold as modulators of cell signaling pathways and potential agents for their use in cancer.
Collapse
Affiliation(s)
- Marco Túlio C Pessôa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del-Rei (UFSJ) Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Jéssica M M Valadares
- Laboratório de Bioquímica Celular, Universidade Federal de São João del-Rei (UFSJ) Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Sayonarah C Rocha
- Laboratório de Bioquímica Celular, Universidade Federal de São João del-Rei (UFSJ) Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Simone C Silva
- Laboratório de Síntese Orgânica e Nanoestruturas, Universidade Federal de São João del-Rei (UFSJ) Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Jeff P McDermott
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center (KUMC), Kansas City, KS, USA
| | - Gladis Sánchez
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center (KUMC), Kansas City, KS, USA
| | - Fernando P Varotti
- Núcleo de Pesquisa em Química Biológica (NQBio), Universidade Federal de São João del-Rei (UFSJ) Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Cristóforo Scavone
- Laboratório de Neurofarmacologia Molecular, Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Rosy I M A Ribeiro
- Laboratório de Patologia Experimental, Universidade Federal de São João del-Rei (UFSJ) Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - José A F P Villar
- Laboratório de Síntese Orgânica e Nanoestruturas, Universidade Federal de São João del-Rei (UFSJ) Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Gustavo Blanco
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center (KUMC), Kansas City, KS, USA
| | - Leandro A Barbosa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del-Rei (UFSJ) Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil.
| |
Collapse
|
26
|
Multipurpose Na + ions mediate excitation and cellular homeostasis: Evolution of the concept of Na + pumps and Na +/Ca 2+ exchangers. Cell Calcium 2020; 87:102166. [PMID: 32006802 DOI: 10.1016/j.ceca.2020.102166] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/14/2022]
Abstract
Ionic signalling is the most ancient form of regulation of cellular functions in response to environmental challenges. Signals, mediated by Na+ fluxes and spatio-temporal fluctuations of Na+ concentration in cellular organelles and cellular compartments contribute to the most fundamental cellular processes such as membrane excitability and energy production. At the very core of ionic signalling lies the Na+-K+ ATP-driven pump (or NKA) which creates trans-plasmalemmal ion gradients that sustain ionic fluxes through ion channels and numerous Na+-dependent transporters that maintain cellular and tissue homeostasis. Here we present a brief account of the history of research into NKA, Na+ -dependent transporters and Na+ signalling.
Collapse
|
27
|
Orlov SN, Tverskoi AM, Sidorenko SV, Smolyaninova LV, Lopina OD, Dulin NO, Klimanova EA. Na,K-ATPase as a target for endogenous cardiotonic steroids: What's the evidence? Genes Dis 2020; 8:259-271. [PMID: 33997173 PMCID: PMC8093582 DOI: 10.1016/j.gendis.2020.01.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/24/2019] [Accepted: 01/09/2020] [Indexed: 12/17/2022] Open
Abstract
With an exception of few reports, the plasma concentration of ouabain and marinobufagenin, mostly studied cardiotonic steroids (CTS) assessed by immunoassay techniques, is less than 1 nM. During the last 3 decades, the implication of these endogenous CTS in the pathogenesis of hypertension and other volume-expanded disorders is widely disputed. The threshold for inhibition by CTS of human and rodent α1-Na,K-ATPase is ∼1 and 1000 nM, respectively, that rules out the functioning of endogenous CTS (ECTS) as natriuretic hormones and regulators of cell adhesion, cell-to-cell communication, gene transcription and translation, which are mediated by dissipation of the transmembrane gradients of monovalent cations. In several types of cells ouabain and marinobufagenin at concentrations corresponding to its plasma level activate Na,K-ATPase, decrease the [Na+]i/[K+]i-ratio and increase cell proliferation. Possible physiological significance and mechanism of non-canonical Na+i/K+i-dependent and Na+i/K+i-independent cell responses to CTS are discussed.
Collapse
Affiliation(s)
- Sergei N Orlov
- MV Lomonosov Moscow State University, Moscow, 119234, Russia.,National Research Tomsk State University, Tomsk, 634050, Russia.,Siberian State Medical University, Tomsk, 634050, Russia
| | | | - Svetlana V Sidorenko
- MV Lomonosov Moscow State University, Moscow, 119234, Russia.,National Research Tomsk State University, Tomsk, 634050, Russia
| | - Larisa V Smolyaninova
- MV Lomonosov Moscow State University, Moscow, 119234, Russia.,National Research Tomsk State University, Tomsk, 634050, Russia
| | - Olga D Lopina
- MV Lomonosov Moscow State University, Moscow, 119234, Russia
| | | | - Elizaveta A Klimanova
- MV Lomonosov Moscow State University, Moscow, 119234, Russia.,National Research Tomsk State University, Tomsk, 634050, Russia
| |
Collapse
|
28
|
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: 54] [Impact Index Per Article: 10.8] [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
|
29
|
Askari A. The other functions of the sodium pump. Cell Calcium 2019; 84:102105. [DOI: 10.1016/j.ceca.2019.102105] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/21/2019] [Accepted: 10/28/2019] [Indexed: 01/14/2023]
|
30
|
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
|
31
|
Penniyaynen VA, Plakhova VB, Rogachevskii IV, Terekhin SG, Podzorova SA, Krylov BV. Molecular mechanisms and signaling by comenic acid in nociceptive neurons influence the pathophysiology of neuropathic pain. ACTA ACUST UNITED AC 2019; 26:245-252. [PMID: 31257013 DOI: 10.1016/j.pathophys.2019.06.003] [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: 03/14/2019] [Revised: 06/10/2019] [Accepted: 06/13/2019] [Indexed: 12/17/2022]
Abstract
Comenic acid (CA), a specific agonist of opioid-like receptors, effectively and safely relieves neuropathic pain by decreasing the NaV1.8 channel voltage sensitivity in the primary sensory neuron membrane. CA triggers downstream signaling cascades, in which the Na,K-ATPase/Src complex plays a key role. After leaving the complex, the signal diverges 'tangentially' and 'radially'. It is directed 'tangentially' along the neuron membrane to NaV1.8 channels, decreasing the effective charge of their activation gating system. In the radial direction moving towards the cell genome, the signal activates the downstream signaling pathway involving PKC and ERK1/2. A remarkable feature of CA is its ability to modulate NaV1.8 channels, which relieves neuropathic pain while simultaneously stimulating neurite growth via the receptor-coupled activation of the ERK1/2-dependent signaling pathway.
Collapse
Affiliation(s)
- Valentina A Penniyaynen
- Laboratory of Physiology of Excitable Membranes, Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Nab. Makarova, 199034, Saint Petersburg, Russia.
| | - Vera B Plakhova
- Laboratory of Physiology of Excitable Membranes, Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Nab. Makarova, 199034, Saint Petersburg, Russia.
| | - Ilya V Rogachevskii
- Laboratory of Physiology of Excitable Membranes, Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Nab. Makarova, 199034, Saint Petersburg, Russia.
| | - Stanislav G Terekhin
- Laboratory of Physiology of Excitable Membranes, Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Nab. Makarova, 199034, Saint Petersburg, Russia.
| | - Svetlana A Podzorova
- Laboratory of Physiology of Excitable Membranes, Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Nab. Makarova, 199034, Saint Petersburg, Russia.
| | - Boris V Krylov
- Laboratory of Physiology of Excitable Membranes, Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Nab. Makarova, 199034, Saint Petersburg, Russia.
| |
Collapse
|
32
|
Li XC, Zheng X, Chen X, Zhao C, Zhu D, Zhang J, Zhuo JL. Genetic and genomic evidence for an important role of the Na +/H + exchanger 3 in blood pressure regulation and angiotensin II-induced hypertension. Physiol Genomics 2019; 51:97-108. [PMID: 30849009 PMCID: PMC6485378 DOI: 10.1152/physiolgenomics.00122.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The sodium (Na+)/hydrogen (H+) exchanger 3 (NHE3) and sodium-potassium adenosine triphosphatase (Na+/K+-ATPase) are two of the most important Na+ transporters in the proximal tubules of the kidney. On the apical membrane side, NHE3 primarily mediates the entry of Na+ into and the exit of H+ from the proximal tubules, directly and indirectly being responsible for reabsorbing ~50% of filtered Na+ in the proximal tubules of the kidney. On the basolateral membrane side, Na+/K+-ATPase serves as a powerful engine driving Na+ out of, while pumping K+ into the proximal tubules against their concentration gradients. While the roles of NHE3 and Na+/K+-ATPase in proximal tubular Na+ transport under in vitro conditions are well recognized, their respective contributions to the basal blood pressure regulation and angiotensin II (ANG II)-induced hypertension remain poorly understood. Recently, we have been fortunate to be able to use genetically modified mouse models with global, kidney- or proximal tubule-specific deletion of NHE3 to directly determine the cause and effect relationship between NHE3, basal blood pressure homeostasis, and ANG II-induced hypertension at the whole body, kidney and/or proximal tubule levels. The purpose of this article is to review the genetic and genomic evidence for an important role of NHE3 with a focus in the regulation of basal blood pressure and ANG II-induced hypertension, as we learned from studies using global, kidney- or proximal tubule-specific NHE3 knockout mice. We hypothesize that NHE3 in the proximal tubules is necessary for maintaining basal blood pressure homeostasis and the development of ANG II-induced hypertension.
Collapse
Affiliation(s)
- Xiao C Li
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Xiaowen Zheng
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Xu Chen
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Chunling Zhao
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Dongmin Zhu
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Jianfeng Zhang
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| | - Jia L Zhuo
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology; Division of Nephrology, Internal Medicine; Cardiovascular and Renal Research Center; The University of Mississippi Medical Center , Jackson, Mississippi
| |
Collapse
|
33
|
Plakhova VB, Penniyaynen VA, Yachnev IL, Rogachevskii IV, Podzorova SA, Krylov BV. Src kinase controls signaling pathways in sensory neuron triggered by low-power infrared radiation. Can J Physiol Pharmacol 2019; 97:400-406. [PMID: 30712368 DOI: 10.1139/cjpp-2018-0602] [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] [Indexed: 11/22/2022]
Abstract
Low-power (non-thermal) infrared (IR) radiation with the wavelength of 10.6 μm activates the Na,K-ATPase transducer function in sensory neurons, which is manifested in decrease of NaV1.8 channel voltage sensitivity at the cellular membrane level and in inhibition of growth of chick embryo dorsal root ganglia neurites at the tissue level. It is shown that the effect of low-power IR radiation is totally blocked by a specific Src kinase inhibitor, PP2. Upon irradiation on the background of PP2, the effective charge of NaV1.8 channel activation gating system does not differ from its control value in patch-clamp experiments, and the area index of sensory ganglia neurites growth remains unchanged as compared with the control in organotypic tissue culture. The data obtained demonstrate that Src kinase is involved in intracellular signaling pathways triggered by CO2 laser low-power IR radiation by the transducer-activated mechanism. This is the first indication that in primary sensory neuron the signals of low-power IR radiation are sensed, amplified, and transduced by the Na,K-ATPase/Src complex and not by G proteins.
Collapse
Affiliation(s)
- Vera B Plakhova
- Laboratory of Physiology of Excitable Membranes, Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Nab. Makarova, 199034, Saint Petersburg, Russia.,Laboratory of Physiology of Excitable Membranes, Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Nab. Makarova, 199034, Saint Petersburg, Russia
| | - Valentina A Penniyaynen
- Laboratory of Physiology of Excitable Membranes, Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Nab. Makarova, 199034, Saint Petersburg, Russia.,Laboratory of Physiology of Excitable Membranes, Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Nab. Makarova, 199034, Saint Petersburg, Russia
| | - Igor L Yachnev
- Laboratory of Physiology of Excitable Membranes, Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Nab. Makarova, 199034, Saint Petersburg, Russia.,Laboratory of Physiology of Excitable Membranes, Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Nab. Makarova, 199034, Saint Petersburg, Russia
| | - Ilya V Rogachevskii
- Laboratory of Physiology of Excitable Membranes, Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Nab. Makarova, 199034, Saint Petersburg, Russia.,Laboratory of Physiology of Excitable Membranes, Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Nab. Makarova, 199034, Saint Petersburg, Russia
| | - Svetlana A Podzorova
- Laboratory of Physiology of Excitable Membranes, Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Nab. Makarova, 199034, Saint Petersburg, Russia.,Laboratory of Physiology of Excitable Membranes, Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Nab. Makarova, 199034, Saint Petersburg, Russia
| | - Boris V Krylov
- Laboratory of Physiology of Excitable Membranes, Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Nab. Makarova, 199034, Saint Petersburg, Russia
| |
Collapse
|
34
|
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
|
35
|
Khalaf FK, Dube P, Mohamed A, Tian J, Malhotra D, Haller ST, Kennedy DJ. Cardiotonic Steroids and the Sodium Trade Balance: New Insights into Trade-Off Mechanisms Mediated by the Na⁺/K⁺-ATPase. Int J Mol Sci 2018; 19:E2576. [PMID: 30200235 PMCID: PMC6165267 DOI: 10.3390/ijms19092576] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 08/24/2018] [Accepted: 08/26/2018] [Indexed: 02/06/2023] Open
Abstract
In 1972 Neal Bricker presented the "trade-off" hypothesis in which he detailed the role of physiological adaptation processes in mediating some of the pathophysiology associated with declines in renal function. In the late 1990's Xie and Askari published seminal studies indicating that the Na⁺/K⁺-ATPase (NKA) was not only an ion pump, but also a signal transducer that interacts with several signaling partners. Since this discovery, numerous studies from multiple laboratories have shown that the NKA is a central player in mediating some of these long-term "trade-offs" of the physiological adaptation processes which Bricker originally proposed in the 1970's. In fact, NKA ligands such as cardiotonic steroids (CTS), have been shown to signal through NKA, and consequently been implicated in mediating both adaptive and maladaptive responses to volume overload such as fibrosis and oxidative stress. In this review we will emphasize the role the NKA plays in this "trade-off" with respect to CTS signaling and its implication in inflammation and fibrosis in target organs including the heart, kidney, and vasculature. As inflammation and fibrosis exhibit key roles in the pathogenesis of a number of clinical disorders such as chronic kidney disease, heart failure, atherosclerosis, obesity, preeclampsia, and aging, this review will also highlight the role of newly discovered NKA signaling partners in mediating some of these conditions.
Collapse
Affiliation(s)
- Fatimah K Khalaf
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Health Education Building RM 205, 3000 Arlington Ave, Toledo, OH 43614, USA.
| | - Prabhatchandra Dube
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Health Education Building RM 205, 3000 Arlington Ave, Toledo, OH 43614, USA.
| | - Amal Mohamed
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Health Education Building RM 205, 3000 Arlington Ave, Toledo, OH 43614, USA.
| | - Jiang Tian
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Health Education Building RM 205, 3000 Arlington Ave, Toledo, OH 43614, USA.
| | - Deepak Malhotra
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Health Education Building RM 205, 3000 Arlington Ave, Toledo, OH 43614, USA.
| | - Steven T Haller
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Health Education Building RM 205, 3000 Arlington Ave, Toledo, OH 43614, USA.
| | - David J Kennedy
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Health Education Building RM 205, 3000 Arlington Ave, Toledo, OH 43614, USA.
| |
Collapse
|
36
|
Fedorova OV, Ishkaraeva VV, Grigorova YN, Reznik VA, Kolodkin NI, Zazerskaya IE, Zernetkina V, Agalakova NI, Tapilskaya NI, Adair CD, Lakatta EG, Bagrov AY. Antibody to Marinobufagenin Reverses Placenta-Induced Fibrosis of Umbilical Arteries in Preeclampsia. Int J Mol Sci 2018; 19:ijms19082377. [PMID: 30104471 PMCID: PMC6121256 DOI: 10.3390/ijms19082377] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/04/2018] [Accepted: 08/07/2018] [Indexed: 02/01/2023] Open
Abstract
Background: Previous studies implicated cardiotonic steroids, including Na/K-ATPase inhibitor marinobufagenin (MBG), in the pathogenesis of preeclampsia (PE). Immunoneutralization of heightened MBG by Digibind, a digoxin antibody, reduces blood pressure (BP) in patients with PE, and anti-MBG monoclonal antibody lessens BP in a rat model of PE. Recently, we demonstrated that MBG induces fibrosis in cardiovascular tissues via a mechanism involving inhibition of Fli-1, a nuclear transcription factor and a negative regulator of collagen-1 synthesis. Objectives and Methods: We hypothesized that in PE, elevated placental MBG levels are associated with development of fibrosis in umbilical arteries. Eleven patients with PE (mean BP 124 ± 4 mmHg; age 29 ± 2 years; 39 weeks gest. age) and 10 gestational age-matched normal pregnant subjects (mean BP 92 ± 2 mmHg; controls) were enrolled in the clinical study. Results: PE was associated with a higher placental (0.04 ± 0.01 vs. 0.49 ± 0.11 pmol/g; p < 0.01) and plasma MBG (0.5 ± 0.1 vs. 1.6 ± 0.5 nmol/L; p < 0.01), lower Na/K-ATPase activity in erythrocytes (2.7 ± 0.2 vs. 1.5 ± 0.2 µmol Pi/mL/hr; p < 0.01), 9-fold decrease of Fli-1 level and 2.5-fold increase of collagen-1 in placentae (p < 0.01) vs. control. Incubation of umbilical arteries from control patients with 1 nmol/L MBG was associated with four-fold decrease in Fli-1 level and two-fold increase in collagen-1 level vs. those incubated with placebo (p < 0.01), i.e., physiological concentration of MBG mimicked effect of PE in vitro. Collagen-1 abundance in umbilical arteries from PE patients was 4-fold higher than in control arteries, and this PE-associated fibrosis was reversed by monoclonal anti-MBG antibody ex vivo. Conclusion: These results demonstrate that elevated placental MBG level is implicated in the development of fibrosis of the placenta and umbilical arteries in PE.
Collapse
Affiliation(s)
- Olga V Fedorova
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA.
| | - Valentina V Ishkaraeva
- Institute of Neonatology, Almazov Federal Heart, Blood and Endocrinology Center, St. Petersburg 197431, Russia.
| | - Yulia N Grigorova
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA.
| | - Vitaly A Reznik
- Department of Obstetrics and Gynecology, School of Pediatric Medicine, St. Petersburg 194353, Russia.
| | - Nikolai I Kolodkin
- Institute of Highly Pure Biopreparations, St. Petersburg 197110, Russia.
- Sechenov Institute of Evolutionary Physiology and Biochemistry, 44 Torez Prospect, St. Petersburg 194223, Russia.
| | - Irina E Zazerskaya
- Institute of Neonatology, Almazov Federal Heart, Blood and Endocrinology Center, St. Petersburg 197431, Russia.
| | - Valentina Zernetkina
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA.
| | - Natalia I Agalakova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, 44 Torez Prospect, St. Petersburg 194223, Russia.
| | | | - C David Adair
- Department of Obstetrics and Gynecology, University of Tennessee, Chattanooga, TN 37403, USA.
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA.
| | - Alexei Y Bagrov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, 44 Torez Prospect, St. Petersburg 194223, Russia.
| |
Collapse
|
37
|
Marck PV, Pierre SV. Na/K-ATPase Signaling and Cardiac Pre/Postconditioning with Cardiotonic Steroids. Int J Mol Sci 2018; 19:ijms19082336. [PMID: 30096873 PMCID: PMC6121447 DOI: 10.3390/ijms19082336] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/05/2018] [Accepted: 08/06/2018] [Indexed: 12/13/2022] Open
Abstract
The first reports of cardiac Na/K-ATPase signaling, published 20 years ago, have opened several major fields of investigations into the cardioprotective action of low/subinotropic concentrations of cardiotonic steroids (CTS). This review focuses on the protective cardiac Na/K-ATPase-mediated signaling triggered by low concentrations of ouabain and other CTS, in the context of the enduring debate over the use of CTS in the ischemic heart. Indeed, as basic and clinical research continues to support effectiveness and feasibility of conditioning interventions against ischemia/reperfusion injury in acute myocardial infarction (AMI), the mechanistic information available to date suggests that unique features of CTS-based conditioning could be highly suitable, alone /or as a combinatory approach.
Collapse
Affiliation(s)
- Pauline V Marck
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, WV 25701, USA.
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, WV 25701, USA.
| |
Collapse
|
38
|
Kutz LC, Mukherji ST, Wang X, Bryant A, Larre I, Heiny JA, Lingrel JB, Pierre SV, Xie Z. Isoform-specific role of Na/K-ATPase α1 in skeletal muscle. Am J Physiol Endocrinol Metab 2018; 314:E620-E629. [PMID: 29438630 PMCID: PMC6032065 DOI: 10.1152/ajpendo.00275.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The distribution of Na/K-ATPase α-isoforms in skeletal muscle is unique, with α1 as the minor (15%) isoform and α2 comprising the bulk of the Na/K-ATPase pool. The acute and isoform-specific role of α2 in muscle performance and resistance to fatigue is well known, but the isoform-specific role of α1 has not been as thoroughly investigated. In vitro, we reported that α1 has a role in promoting cell growth that is not supported by α2. To assess whether α1 serves this isoform-specific trophic role in the skeletal muscle, we used Na/K-ATPase α1-haploinsufficient (α1+/-) mice. A 30% decrease of Na/K-ATPase α1 protein expression without change in α2 induced a modest yet significant decrease of 10% weight in the oxidative soleus muscle. In contrast, the mixed plantaris and glycolytic extensor digitorum longus weights were not significantly affected, likely because of their very low expression level of α1 compared with the soleus. The soleus mass reduction occurred without change in total Na/K-ATPase activity or glycogen metabolism. Serum analytes including K+, fat tissue mass, and exercise capacity were not altered in α1+/- mice. The impact of α1 content on soleus muscle mass is consistent with a Na/K-ATPase α1-specific role in skeletal muscle growth that cannot be fulfilled by α2. The preserved running capacity in α1+/- is in sharp contrast with previously reported consequences of genetic manipulation of α2. Taken together, these results lend further support to the concept of distinct isoform-specific functions of Na/K-ATPase α1 and α2 in skeletal muscle.
Collapse
Affiliation(s)
- Laura C Kutz
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Shreya T Mukherji
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Xiaoliang Wang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Amber Bryant
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Isabel Larre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Judith A Heiny
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine , Cincinnati, Ohio
| | - Jerry B Lingrel
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine , Cincinnati, Ohio
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| |
Collapse
|
39
|
Zhang C, Wang H, Chen Z, Zhuang L, Xu L, Ning Z, Zhu Z, Wang P, Meng Z. Carbonic anhydrase 2 inhibits epithelial–mesenchymal transition and metastasis in hepatocellular carcinoma. Carcinogenesis 2018; 39:562-570. [DOI: 10.1093/carcin/bgx148] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Chenyue Zhang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Haiyong Wang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, China
| | - Zhiao Chen
- Fudan University Shanghai Cancer Center and Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liping Zhuang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Litao Xu
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhouyu Ning
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhenfeng Zhu
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peng Wang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiqiang Meng
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
40
|
Lopachev AV, Abaimov DA, Fedorova TN, Lopacheva OM, Akkuratova NV, Akkuratov EE. Cardiotonic Steroids as Potential Endogenous Regulators in the Nervous System. NEUROCHEM J+ 2018. [DOI: 10.1134/s1819712418010087] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
41
|
Wong RW, Lingwood CA, Ostrowski MA, Cabral T, Cochrane A. Cardiac glycoside/aglycones inhibit HIV-1 gene expression by a mechanism requiring MEK1/2-ERK1/2 signaling. Sci Rep 2018; 8:850. [PMID: 29339801 PMCID: PMC5770468 DOI: 10.1038/s41598-018-19298-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 11/07/2017] [Indexed: 12/14/2022] Open
Abstract
The capacity of HIV-1 to develop resistance to current drugs calls for innovative strategies to control this infection. We aimed at developing novel inhibitors of HIV-1 replication by targeting viral RNA processing—a stage dependent on conserved host processes. We previously reported that digoxin is a potent inhibitor of this stage. Herein, we identify 12 other cardiac glycoside/aglycones or cardiotonic steroids (CSs) that impede HIV growth in HIV-infected T cells from clinical patients at IC50s (1.1–1.3 nM) that are 2–26 times below concentrations used in patients with heart conditions. We subsequently demonstrate that CSs inhibit HIV-1 gene expression in part through modulation of MEK1/2-ERK1/2 signaling via interaction with the Na+/K+-ATPase, independent of alterations in intracellular Ca2+. Supporting this hypothesis, depletion of the Na+/K+-ATPase or addition of a MEK1/2-ERK1/2 activator also impairs HIV-1 gene expression. Similar to digoxin, all CSs tested induce oversplicing of HIV-1 RNAs, reducing unspliced (Gag) and singly spliced RNAs (Env/p14-Tat) encoding essential HIV-1 structural/regulatory proteins. Furthermore, all CSs cause nuclear retention of genomic/unspliced RNAs, supporting viral RNA processing as the underlying mechanism for their disruption of HIV-1 replication. These findings call for further in vivo validation and supports the targeting of cellular processes to control HIV-1 infection.
Collapse
Affiliation(s)
- Raymond W Wong
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Clifford A Lingwood
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S1A8, Canada.,Division of Molecular Structure and Function, Hospital for Sick Children, Toronto, ON, M5G1X8, Canada.,Department of Biochemistry, University of Toronto, Toronto, Ontario, M5S1A8, Canada
| | - Mario A Ostrowski
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital Toronto, Toronto, ON, M5B1W8, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, M5S1A8, Canada.,Department of Immunology, University of Toronto, Toronto, ON, M5S1A8, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Tyler Cabral
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Alan Cochrane
- Institute of Medical Science, University of Toronto, Toronto, ON, M5S1A8, Canada. .,Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada.
| |
Collapse
|
42
|
Pessôa MTC, Alves SLG, Taranto AG, Villar JAFP, Blanco G, Barbosa LA. Selectivity analyses of γ-benzylidene digoxin derivatives to different Na,K-ATPase α isoforms: a molecular docking approach. J Enzyme Inhib Med Chem 2017; 33:85-97. [PMID: 29115894 PMCID: PMC6009882 DOI: 10.1080/14756366.2017.1380637] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Digoxin and other cardiotonic steroids (CTS) exert their effect by inhibiting Na,K-ATPase (NKA) activity. CTS bind to the various NKA isoforms that are expressed in different cell types, which gives CTS their narrow therapeutic index. We have synthesised a series of digoxin derivatives (γ-Benzylidene digoxin derivatives) with substitutions in the lactone ring (including non-oxygen and ether groups), to obtain CTS with better NKA isoform specificity. Some of these derivatives show some NKA isoform selective effects, with BD-3, BD-8, and BD-13 increasing NKA α2 activity, BD-5 inhibiting NKA α1 and NKA α3, BD-10 reducing NKA α1, but stimulating NKA α2 and α3; and BD-14, BD-15, and BD-16 enhancing NKA α3 activity. A molecular-docking approach favoured NKA isoform specific interactions for the compounds that supported their observed activity. These results show that BD compounds are a new type of CTS with the capacity to target NKA activity in an isoform-specific manner.
Collapse
Affiliation(s)
- Marco T C Pessôa
- a Laboratório de Bioquímica Celular , Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú , Divinópolis , Brazil
| | - Silmara L G Alves
- b Laboratório de Síntese Orgânica e Nanoestruturas , Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú , Divinópolis , Brazil
| | - Alex G Taranto
- c Laboratório de Modelagem Molecular , Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú , Divinópolis , Brazil
| | - José A F P Villar
- b Laboratório de Síntese Orgânica e Nanoestruturas , Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú , Divinópolis , Brazil
| | - Gustavo Blanco
- d Department of Molecular and Integrative Physiology , Kansas University Medical Center , Kansas City , KS , USA
| | - Leandro A Barbosa
- a Laboratório de Bioquímica Celular , Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú , Divinópolis , Brazil
| |
Collapse
|
43
|
Blaustein MP. The pump, the exchanger, and the holy spirit: origins and 40-year evolution of ideas about the ouabain-Na + pump endocrine system. Am J Physiol Cell Physiol 2017; 314:C3-C26. [PMID: 28971835 DOI: 10.1152/ajpcell.00196.2017] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Two prescient 1953 publications set the stage for the elucidation of a novel endocrine system: Schatzmann's report that cardiotonic steroids (CTSs) are all Na+ pump inhibitors, and Szent-Gyorgi's suggestion that there is an endogenous "missing screw" in heart failure that CTSs like digoxin may replace. In 1977 I postulated that an endogenous Na+ pump inhibitor acts as a natriuretic hormone and simultaneously elevates blood pressure (BP) in salt-dependent hypertension. This hypothesis was based on the idea that excess renal salt retention promoted the secretion of a CTS-like hormone that inhibits renal Na+ pumps and salt reabsorption. The hormone also inhibits arterial Na+ pumps, elevates myocyte Na+ and promotes Na/Ca exchanger-mediated Ca2+ gain. This enhances vasoconstriction and arterial tone-the hallmark of hypertension. Here I describe how those ideas led to the discovery that the CTS-like hormone is endogenous ouabain (EO), a key factor in the pathogenesis of hypertension and heart failure. Seminal observations that underlie the still-emerging picture of the EO-Na+ pump endocrine system in the physiology and pathophysiology of multiple organ systems are summarized. Milestones include: 1) cloning the Na+ pump isoforms and physiological studies of mutated pumps in mice; 2) discovery that Na+ pumps are also EO-triggered signaling molecules; 3) demonstration that ouabain, but not digoxin, is hypertensinogenic; 4) elucidation of EO's roles in kidney development and cardiovascular and renal physiology and pathophysiology; 5) discovery of "brain ouabain", a component of a novel hypothalamic neuromodulatory pathway; and 6) finding that EO and its brain receptors modulate behavior and learning.
Collapse
Affiliation(s)
- Mordecai P Blaustein
- Departments of Physiology and Medicine, University of Maryland School of Medicine , Baltimore, Maryland
| |
Collapse
|
44
|
Dey S, Krishna S, Anthony NB, Rhoads DD. Further investigation of a quantitative trait locus for ascites on chromosome 9 in broiler chicken lines. Poult Sci 2017; 96:788-797. [PMID: 28339549 DOI: 10.3382/ps/pew380] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/20/2016] [Indexed: 01/05/2023] Open
Abstract
Previously, we reported a genome wide association study (GWAS) that had shown association of a region between 11.8 and 13.6 Mbp on chromosome 9 with ascites phenotype in broilers. We had used microsatellite loci to demonstrate an association of particular genotypes for this region with ascites in experimental ascites lines and commercial broiler breeder lines. We identified two potential candidate genes, AGTR1 and UTS2D, within that chromosomal region for mediating the quantitative effect. We have now extended our analysis using SNPs for these genes to assess association with resistance or susceptibility to ascites in these same broiler lines. Surprisingly, in contrast to our previous GWAS and microsatellite data for this region, we find no association of the SNP genotypes or haplotypes in the region suggesting that the two genes might have limited association with the disease phenotype.
Collapse
Affiliation(s)
- Shatovisha Dey
- Program in Cell and Molecular Biology, University of Arkansas, Fayetteville, AR 72701.,Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701
| | - Sriram Krishna
- Program in Cell and Molecular Biology, University of Arkansas, Fayetteville, AR 72701
| | - Nicholas B Anthony
- Program in Cell and Molecular Biology, University of Arkansas, Fayetteville, AR 72701.,Department of Poultry Sciences, University of Arkansas, Fayetteville, AR 72701
| | - Douglas D Rhoads
- Program in Cell and Molecular Biology, University of Arkansas, Fayetteville, AR 72701.,Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701
| |
Collapse
|
45
|
Ouabain affects cell migration via Na,K-ATPase-p130cas and via nucleus-centrosome association. PLoS One 2017; 12:e0183343. [PMID: 28817661 PMCID: PMC5560699 DOI: 10.1371/journal.pone.0183343] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/02/2017] [Indexed: 01/12/2023] Open
Abstract
Na,K-ATPase is a membrane protein that catalyzes ATP to maintain transmembrane sodium and potassium gradients. In addition, Na,K-ATPase also acts as a signal-transducing receptor for cardiotonic steroids such as ouabain and activates a number of signalling pathways. Several studies report that ouabain affects cell migration. Here we used ouabain at concentrations far below those required to block Na,K-ATPase pump activity and show that it significantly reduced RPE cell migration through two mechanisms. It causes dephosphorylation of a 130 kD protein, which we identify as p130cas. Src is involved, because Src inhibitors, but not inhibitors of other kinases tested, caused a similar reduction in p130cas phosphorylation and ouabain increased the association of Na,K-ATPase and Src. Knockdown of p130cas by siRNA reduced cell migration. Unexpectedly, ouabain induced separation of nucleus and centrosome, also leading to a block in cell migration. Inhibitor and siRNA experiments show that this effect is mediated by ERK1,2. This is the first report showing that ouabain can regulate cell migration by affecting nucleus-centrosome association.
Collapse
|
46
|
Cui X, Xie Z. Protein Interaction and Na/K-ATPase-Mediated Signal Transduction. Molecules 2017; 22:molecules22060990. [PMID: 28613263 PMCID: PMC6152704 DOI: 10.3390/molecules22060990] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/01/2017] [Accepted: 06/02/2017] [Indexed: 02/05/2023] Open
Abstract
The Na/K-ATPase (NKA), or Na pump, is a member of the P-type ATPase superfamily. In addition to pumping ions across cell membrane, it is engaged in assembly of multiple protein complexes in the plasma membrane. This assembly allows NKA to perform many non-pumping functions including signal transduction that are important for animal physiology and disease progression. This article will focus on the role of protein interaction in NKA-mediated signal transduction, and its potential utility as target for developing new therapeutics.
Collapse
Affiliation(s)
- Xiaoyu Cui
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA.
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA.
| |
Collapse
|
47
|
On the Many Actions of Ouabain: Pro-Cystogenic Effects in Autosomal Dominant Polycystic Kidney Disease. Molecules 2017; 22:molecules22050729. [PMID: 28467389 PMCID: PMC5688955 DOI: 10.3390/molecules22050729] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/19/2017] [Accepted: 04/30/2017] [Indexed: 02/07/2023] Open
Abstract
Ouabain and other cardenolides are steroidal compounds originally discovered in plants. Cardenolides were first used as poisons, but after finding their beneficial cardiotonic effects, they were rapidly included in the medical pharmacopeia. The use of cardenolides to treat congestive heart failure remained empirical for centuries and only relatively recently, their mechanisms of action became better understood. A breakthrough came with the discovery that ouabain and other cardenolides exist as endogenous compounds that circulate in the bloodstream of mammals. This elevated these compounds to the category of hormones and opened new lines of investigation directed to further study their biological role. Another important discovery was the finding that the effect of ouabain was mediated not only by inhibition of the activity of the Na,K-ATPase (NKA), but by the unexpected role of NKA as a receptor and a signal transducer, which activates a complex cascade of intracellular second messengers in the cell. This broadened the interest for ouabain and showed that it exerts actions that go beyond its cardiotonic effect. It is now clear that ouabain regulates multiple cell functions, including cell proliferation and hypertrophy, apoptosis, cell adhesion, cell migration, and cell metabolism in a cell and tissue type specific manner. This review article focuses on the cardenolide ouabain and discusses its various in vitro and in vivo effects, its role as an endogenous compound, its mechanisms of action, and its potential use as a therapeutic agent; placing especial emphasis on our findings of ouabain as a pro-cystogenic agent in autosomal dominant polycystic kidney disease (ADPKD).
Collapse
|
48
|
Wang X, Liu J, Drummond CA, Shapiro JI. Sodium potassium adenosine triphosphatase (Na/K-ATPase) as a therapeutic target for uremic cardiomyopathy. Expert Opin Ther Targets 2017; 21:531-541. [PMID: 28338377 PMCID: PMC5590225 DOI: 10.1080/14728222.2017.1311864] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 03/23/2017] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Clinically, patients with significant reductions in renal function present with cardiovascular dysfunction typically termed, uremic cardiomyopathy. It is a progressive series of cardiac pathophysiological changes, including left ventricular diastolic dysfunction and hypertrophy (LVH) which sometimes progress to left ventricular dilation (LVD) and systolic dysfunction in the setting of chronic kidney disease (CKD). Uremic cardiomyopathy is almost ubiquitous in patients afflicted with end stage renal disease (ESRD). Areas covered: This article reviews recent epidemiology, pathophysiology of uremic cardiomyopathy and provide a board overview of Na/K-ATPase research with detailed discussion on the mechanisms of Na/K-ATPase/Src/ROS amplification loop. We also present clinical and preclinical evidences as well as molecular mechanism of this amplification loop in the development of uremic cardiomyopathy. A potential therapeutic peptide that targets on this loop is discussed. Expert opinion: Current clinical treatment for uremic cardiomyopathy remains disappointing. Targeting the ROS amplification loop mediated by the Na/K-ATPase signaling function may provide a novel therapeutic target for uremic cardiomyopathy and related diseases. Additional studies of Na/K-ATPase and other strategies that regulate this loop will lead to new therapeutics.
Collapse
Affiliation(s)
- Xiaoliang Wang
- a Joan C Edwards School of Medicine at Marshall University , Huntington , WV , United States
- b University of Toledo College of Medicine and Life Sciences , Toledo , OH , United States
| | - Jiang Liu
- a Joan C Edwards School of Medicine at Marshall University , Huntington , WV , United States
| | - Christopher A Drummond
- b University of Toledo College of Medicine and Life Sciences , Toledo , OH , United States
| | - Joseph I Shapiro
- a Joan C Edwards School of Medicine at Marshall University , Huntington , WV , United States
| |
Collapse
|
49
|
Sahoo SS, Mishra C, Rout M, Nayak G, Mohanty ST, Panigrahy KK. Comparative in silico and protein-protein interaction network analysis of ATP1A1 gene. GENE REPORTS 2016. [DOI: 10.1016/j.genrep.2016.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
50
|
Bai Y, Wu J, Li D, Morgan EE, Liu J, Zhao X, Walsh A, Saikumar J, Tinkel J, Joe B, Gupta R, Liu L. Differential roles of caveolin-1 in ouabain-induced Na+/K+-ATPase cardiac signaling and contractility. Physiol Genomics 2016; 48:739-748. [PMID: 27519543 PMCID: PMC5243228 DOI: 10.1152/physiolgenomics.00042.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 08/03/2016] [Indexed: 11/22/2022] Open
Abstract
Binding of ouabain to cardiac Na+/K+-ATPase initiates cell signaling and causes contractility in cardiomyocytes. It is widely accepted that caveolins, structural proteins of caveolae, have been implicated in signal transduction. It is known that caveolae play a role in Na+/K+-ATPase functions. Regulation of caveolin-1 in ouabain-mediated cardiac signaling and contractility has never been reported. The aim of this study is to compare ouabain-induced cardiac signaling and contractility in wild-type (WT) and caveolin-1 knockout (cav-1 KO) mice. In contrast with WT cardiomyocytes, ouabain-induced signaling e.g., activation of phosphoinositide 3-kinase-α/Akt and extracellular signal-regulated kinases (ERK)1/2, and hypertrophic growth were significantly reduced in cav-1 KO cardiomyocytes. Interactions of the Na+/K+-ATPase α1-subunit with caveolin-3 and the Na+/K+-ATPase α1-subunit with PI3K-α were also decreased in cav-1 KO cardiomyocytes. The results from cav-1 KO mouse embryonic fibroblasts also proved that cav-1 significantly attenuated ouabain-induced ERK1/2 activation without alteration in protein and cholesterol distribution in caveolae/lipid rafts. Intriguingly, the effect of ouabain induced positive inotropy in vivo (via transient infusion of ouabain, 0.48 nmol/g body wt) was not attenuated in cav-1 KO mice. Furthermore, ouabain (1-100 μM) induced dose-dependent contractility in isolated working hearts from WT and cav-1 KO mice. The effects of ouabain on contractility between WT and cav-1 KO mice were not significantly different. These results demonstrated differential roles of cav-1 in the regulation of ouabain signaling and contractility. Signaling by ouabain, in contrast to contractility, may be a redundant property of Na+/K+-ATPase.
Collapse
Affiliation(s)
- Yan Bai
- Department of Biochemistry and Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio; Pediatrics Department of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China
| | - Jian Wu
- Department of Biochemistry and Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Daxiang Li
- Department of Biochemistry and Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio; State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, Anhui, China; and
| | - Eric E Morgan
- Center for Hypertension and Personalized Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Jiang Liu
- Department of Pharmacology, Physiology and Toxicology, JCE School of Medicine, Marshall University, Huntington, West Virginia
| | - Xiaochen Zhao
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Aaron Walsh
- Department of Biochemistry and Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Jagannath Saikumar
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Jodi Tinkel
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Bina Joe
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio; Center for Hypertension and Personalized Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Rajesh Gupta
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Lijun Liu
- Department of Biochemistry and Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio; Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio; Center for Hypertension and Personalized Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio;
| |
Collapse
|