1
|
Trant J, Sanchez G, McDermott JP, Blanco G. The cystogenic effects of ouabain in autosomal dominant polycystic kidney disease require cell caveolae. Exp Cell Res 2025; 444:114356. [PMID: 39586486 DOI: 10.1016/j.yexcr.2024.114356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
We have previously shown that the hormone ouabain is a circulating factor which can accelerate the progression of autosomal dominant polycystic kidney disease (ADPKD). At physiologic concentrations, ouabain increases cyst area and fibrosis in kidneys from ADPKD but not wildtype mice. These effects are due to an increased affinity for ouabain by its receptor, Na,K-ATPase (NKA), in the kidneys of ADPKD mice which leads to over-activation of NKA signaling function. Previous studies suggested that ouabain's stimulation of NKA signal transduction is mediated by NKA located within cell caveolae. Here, we determined whether caveolae are involved in the ouabain-induced progression of ADPKD cysts. We generated an ADPKD mouse with a global knockout of the main structural component of caveolae, caveolin-1 (CAV1), which we confirmed lacks caveolae in the kidney. When given physiological amounts of ouabain for 5 months, Pkd1RC/RCCav1-/- mice did not exhibit any changes in cyst progression, contrasting with the Pkd1RC/RC mice which showed a significant increase in cystic area and kidney fibrosis. Also, measures of ouabain-induced cell proliferation, including the number of Ki67-positive nuclei and phosphorylation of the extracellular regulated kinase (ERK) and protein kinase B (Akt), did not increase in the Pkd1RC/RCCav1-/- mice compared with the Pkd1RC/RC mice. Moreover, the abnormally increased affinity for ouabain of NKA in Pkd1RC/RC mice was restored to wildtype levels in the Pkd1RC/RCCav1-/- mice. This work highlights the role of caveolae in ouabain-induced NKA signaling and ADPKD cyst progression.
Collapse
Affiliation(s)
- Jordan Trant
- Department of Cell Biology and Physiology and the Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Gladis Sanchez
- Department of Cell Biology and Physiology and the Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jeffery P McDermott
- Department of Cell Biology and Physiology and the Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Gustavo Blanco
- Department of Cell Biology and Physiology and the Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
2
|
An Z, Tian J, Zhao X, Liu L, Yang X, Zhang M, Zhang L, Song X. Regulation of cardiovascular and cardiac functions by caveolins. FEBS J 2024; 291:3753-3761. [PMID: 37060249 DOI: 10.1111/febs.16798] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/06/2023] [Accepted: 04/13/2023] [Indexed: 04/16/2023]
Abstract
Caveolae are intracellular vesicles with diameters ranging from 50 to 100 nm. The role of caveolins in mediating oxidative stress, autophagy, apoptosis, fibrosis, and vascular remodeling has attracted increasing attention in cardiovascular therapy. Several studies have suggested that caveolin could be a therapeutic target for the treatment of cardiac and/or vascular injury via several pathophysiological mechanisms. Despite substantial advances in our understanding of the basic biology of vesicles over the past decade, the relevance and specific role of these mechanisms in cardiovascular homeostasis remains ambiguous. Here, we review the macroscopic role of caveolins in protecting cardiac function and, at the microscopic level, examine possible cardioprotective caveolar mechanisms, including their antioxidative stress, antiapoptosis, autophagy-regulatory, antifibrosis, and angiogenesis-promoting properties. We believe that the role of caveolins in cardiac functioning has not been fully elucidated and is an important line of future research with several cardioprotective implications.
Collapse
Affiliation(s)
- Ziyu An
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jinfan Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Libo Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Xueyao Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Mingduo Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lijun Zhang
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Huang M, Wang X, Chen Y, Pessoa MT, Terrell KC, Zhang J, Tian J, Xie Z, Pierre SV, Cai L. Role of Na/K-ATPase α1 caveolin-binding motif in adipogenesis. Am J Physiol Cell Physiol 2024; 327:C48-C64. [PMID: 38708522 PMCID: PMC11371328 DOI: 10.1152/ajpcell.00168.2024] [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: 03/14/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024]
Abstract
Deficiencies in mice and in humans have brought to the fore the importance of the caveolar network in key aspects of adipocyte biology. The conserved N-terminal caveolin-binding motif (CBM) of the ubiquitous Na/K-ATPase (NKA) α1 isoform, which allows NKA/caveolin-1 (Cav1) interaction, influences NKA signaling and caveolar distribution. It has been shown to be critical for animal development and ontogenesis, as well as lineage-specific differentiation of human induced pluripotent stem cells (hiPSCs). However, its role in postnatal adipogenesis has not been fully examined. Using a genetic approach to alter CBM in hiPSC-derived adipocytes (iAdi-mCBM) and in mice (mCBM), we investigated the regulatory function of NKA CBM signaling in adipogenesis. Seahorse XF cell metabolism analyses revealed impaired glycolysis and decreased ATP synthesis-coupled respiration in iAdi-mCBM. These metabolic dysfunctions were accompanied by evidence of extensive remodeling of the extracellular matrix (ECM), including increased collagen staining, overexpression of ECM marker genes, and heightened TGF-β signaling uncovered by RNAseq analysis. Rescue of mCBM by lentiviral delivery of WT NKA α1 or treatment of mCBM hiPSCs with the TGF-β inhibitor SB431542 normalized ECM, suggesting that NKA CBM signaling integrity is required for adequate control of TGF-β signaling and ECM stiffness during adipogenesis. The physiological impact was revealed in mCBM male mice with reduced fat mass accompanied by histological and transcriptional evidence of elevated adipose fibrosis and decreased adipocyte size. Based on these findings, we propose that the genetic alteration of the NKA/Cav1 regulatory path uncovered in human iAdi leads to lipodystrophy in mice.NEW & NOTEWORTHY A Na/K-ATPase α1 caveolin-binding motif regulates adipogenesis. Mutation of this binding motif in the mouse leads to reduced fat with increased extracellular matrix production and inflammation. RNA-seq analysis and pharmacological interventions in human iPSC-derived adipocytes revealed that TGF-β signal, rather than Na/K-ATPase-mediated ion transport, is a key mediator of NKA regulation of adipogenesis.
Collapse
Affiliation(s)
- Minqi Huang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Xiaoliang Wang
- Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Yiliang Chen
- Versiti Blood Research Institute, Milwaukee, West Virginia, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Marco T Pessoa
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Kayleigh C Terrell
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Jue Zhang
- Versiti Blood Research Institute, Milwaukee, West Virginia, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Jiang Tian
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
- Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Liquan Cai
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| |
Collapse
|
4
|
Wu Y, Peng W, Chen S, Zeng X, Zhu J, Zhu P. CAV1 Protein Encapsulated in Mouse BMSC-Derived Extracellular Vesicles Alleviates Myocardial Fibrosis Following Myocardial Infarction by Blocking the TGF-β1/SMAD2/c-JUN Axis. J Cardiovasc Transl Res 2024; 17:523-539. [PMID: 38092988 DOI: 10.1007/s12265-023-10472-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/27/2023] [Indexed: 07/03/2024]
Abstract
Extracellular vesicles (EVs) derived from mouse bone marrow mesenchymal stem cells (mBMSCs) convey the CAV1 protein, influencing the TGF-β1/SMAD2/c-JUN pathway and thus the molecular mechanisms underlying myocardial fibrosis (MF) post-myocardial infarction (MI). Through various experimental methods, including transmission electron microscopy, Nanosight analysis, Western blot, ELISA, and qRT-PCR, we isolated, purified, and identified EVs originating from mBMSCs. Bioinformatics and experimental findings show a reduced expression of CAV1 in myocardial fibrosis tissue. Furthermore, our findings suggest that mBMSC-EVs can deliver CAV1 to cardiac fibroblasts (CFs) and that silencing CAV1 in mBMSC-EVs promotes CF fibrosis. In vivo studies further corroborated these findings. In conclusion, mBMSC-EVs mitigate myocardial fibrosis in MI mice by delivering the CAV1 protein, inhibiting the TGF-β1/SMAD2/c-JUN pathway.
Collapse
Affiliation(s)
- Yijin Wu
- Department of Cardiovascular Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No.106 Zhongshan Er Road, Yuexiu District, Guangzhou, 510100, China
| | - Wenying Peng
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, China
| | - Siyao Chen
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, China
| | - Xiaodong Zeng
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, China
| | - Jiade Zhu
- Department of Cardiovascular Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No.106 Zhongshan Er Road, Yuexiu District, Guangzhou, 510100, China
| | - Ping Zhu
- Department of Cardiovascular Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No.106 Zhongshan Er Road, Yuexiu District, Guangzhou, 510100, China.
| |
Collapse
|
5
|
Leite JA, Pôças E, Maia GS, Barbosa L, Quintas LEM, Kawamoto EM, da Silva MLC, Scavone C, de Carvalho LED. Effect of ouabain on calcium signaling in rodent brain: A systematic review of in vitro studies. Front Pharmacol 2022; 13:916312. [PMID: 36105192 PMCID: PMC9465813 DOI: 10.3389/fphar.2022.916312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 07/20/2022] [Indexed: 11/26/2022] Open
Abstract
The Na+/K+-ATPase is an integral membrane ion pump, essential to maintaining osmotic balance in cells in the presence of cardiotonic steroids; more specifically, ouabain can be an endogenous modulator of the Na+/K+-ATPase. Here, we conducted a systematic review of the in vitro effects of cardiotonic steroids on Ca2+ in the brain of rats and mice. Methods: The review was carried out using the PubMed, Virtual Health Library, and EMBASE databases (between 12 June 2020 and 30 June 2020) and followed the guidelines described in the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA). Results: in total, 829 references were identified in the electronic databases; however, only 20 articles were considered, on the basis of the inclusion criteria. The studies demonstrated the effects of ouabain on Ca2+ signaling in synaptosomes, brain slices, and cultures of rat and mouse cells. In addition to the well-known cytotoxic effects of high doses of ouabain, resulting from indirect stimulation of the reverse mode of the Na+/Ca2+ exchanger and increased intracellular Ca2+, other effects have been reported. Ouabain-mediated Ca2+ signaling was able to act increasing cholinergic, noradrenergic and glutamatergic neurotransmission. Furthermore, ouabain significantly increased intracellular signaling molecules such as InsPs, IP3 and cAMP. Moreover treatment with low doses of ouabain stimulated myelin basic protein synthesis. Ouabain-induced intracellular Ca2+ increase may promote the activation of important cell signaling pathways involved in cellular homeostasis and function. Thus, the study of the application of ouabain in low doses being promising for application in neurological diseases. Systematic Review Registration:https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42020204498, identifier CRD42020204498.
Collapse
Affiliation(s)
- Jacqueline Alves Leite
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Elisa Pôças
- Campus Realengo, Instituto Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gisele Silva Maia
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, São Paulo, Brazil
| | - Leandro Barbosa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, São Paulo, Brazil
| | - Luis Eduardo M. Quintas
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elisa Mitiko Kawamoto
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | | | - Cristoforo Scavone
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Luciana E. Drumond de Carvalho
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, São Paulo, Brazil
- *Correspondence: Luciana E. Drumond de Carvalho,
| |
Collapse
|
6
|
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: 12] [Impact Index Per Article: 3.0] [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
|
7
|
Silva LND, Garcia IJP, Valadares JMM, Pessoa MTC, Toledo MM, Machado MV, Busch MS, Rocha I, Villar JAFP, Atella GC, Santos HL, Cortes VF, Barbosa LA. Evaluation of Cardiotonic Steroid Modulation of Cellular Cholesterol and Phospholipid. J Membr Biol 2021; 254:499-512. [PMID: 34716469 DOI: 10.1007/s00232-021-00203-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 10/09/2021] [Indexed: 02/05/2023]
Abstract
We have previously shown that 21-benzylidene digoxin (21-BD) increases the total cholesterol and phospholipid content on the membrane of HeLa cells. Lipid modulation caused by cardiotonic steroids (CTS) is still unexplored. Therefore, the aim of the present study was to evaluate the cholesterol and phospholipid modulation of the cell membrane caused by ouabain and 21-BD and the possible involvement of the caveolae on this modulation. For this, one cell line containing caveolae (HeLa) and other not containing (Caco-2) were used. The modulation of the lipid profile was evaluated by total cholesterol and phospholipids measurements, and identification of membrane phospholipids by HPTLC. The cholesterol distribution was evaluated by filipin staining. The caveolin-1 expression was evaluated by Western Blotting. Ouabain had no effect on the total membrane lipid content in both cell lines. However, 21-BD increased total membrane phospholipid content and had no effect on the membrane cholesterol content in Caco-2 cells. CTS were not able to alter the specific phospholipids content. In the filipin experiments, 21-BD provoked a remarkable redistribution of cholesterol to the perinuclear region of HeLa cells. In Caco-2 cells, it was observed only a slight increase in cholesterol, especially as intracellular vesicles. The caveolin-1 expression was not altered by any of the compounds. Our data mainly show different effects of two cardiotonic steroids. Ouabain had no effect on the lipid profile of cells, whereas 21-BD causes important changes in cholesterol and phospholipid content. Therefore, the modulation of cholesterol content in the plasma membrane of HeLa cells is not correlated with the expression of caveolin-1.
Collapse
Affiliation(s)
- Lilian N D Silva
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Divinópolis, MG, Zip Code: 35501-296, Brazil.,Laboratório de Bioquímica de Membranas e ATPases, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Israel J P Garcia
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Divinópolis, MG, Zip Code: 35501-296, Brazil.,Laboratório de Bioquímica de Membranas e ATPases, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Jessica M M Valadares
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Divinópolis, MG, Zip Code: 35501-296, Brazil.,Laboratório de Bioquímica de Membranas e ATPases, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Marco Tulio C Pessoa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Divinópolis, MG, Zip Code: 35501-296, Brazil.,Laboratório de Bioquímica de Membranas e ATPases, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Marina Marques Toledo
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Divinópolis, MG, Zip Code: 35501-296, Brazil.,Laboratório de Bioquímica de Membranas e ATPases, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Matheus V Machado
- Laboratório de Síntese Orgânica e Nanoestruturas, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Mileane Souza Busch
- Laboratório de Bioquímica de Lipídios, Instituto de Bioquímica Médica Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Isabella Rocha
- Laboratório de Bioquímica de Lipídios, Instituto de Bioquímica Médica Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - José Augusto F P Villar
- Laboratório de Síntese Orgânica e Nanoestruturas, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Georgia C Atella
- Laboratório de Bioquímica de Lipídios, Instituto de Bioquímica Médica Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Herica L Santos
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Divinópolis, MG, Zip Code: 35501-296, Brazil.,Laboratório de Bioquímica de Membranas e ATPases, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Vanessa F Cortes
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Divinópolis, MG, Zip Code: 35501-296, Brazil. .,Laboratório de Bioquímica de Membranas e ATPases, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil.
| | - Leandro A Barbosa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Divinópolis, MG, Zip Code: 35501-296, Brazil. .,Laboratório de Bioquímica de Membranas e ATPases, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil.
| |
Collapse
|
8
|
Zhang J, Li X, Yu H, Larre I, Dube PR, Kennedy DJ, Tang WHW, Westfall K, Pierre SV, Xie Z, Chen Y. Regulation of Na/K-ATPase expression by cholesterol: isoform specificity and the molecular mechanism. Am J Physiol Cell Physiol 2020; 319:C1107-C1119. [PMID: 32997514 DOI: 10.1152/ajpcell.00083.2020] [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] [Indexed: 11/22/2022]
Abstract
We have reported that the reduction in plasma membrane cholesterol could decrease cellular Na/K-ATPase α1-expression through a Src-dependent pathway. However, it is unclear whether cholesterol could regulate other Na/K-ATPase α-isoforms and the molecular mechanisms of this regulation are not fully understood. Here we used cells expressing different Na/K-ATPase α isoforms and found that membrane cholesterol reduction by U18666A decreased expression of the α1-isoform but not the α2- or α3-isoform. Imaging analyses showed the cellular redistribution of α1 and α3 but not α2. Moreover, U18666A led to redistribution of α1 to late endosomes/lysosomes, while the proteasome inhibitor blocked α1-reduction by U18666A. These results suggest that the regulation of the Na/K-ATPase α-subunit by cholesterol is isoform specific and α1 is unique in this regulation through the endocytosis-proteasome pathway. Mechanistically, loss-of-Src binding mutation of A425P in α1 lost its capacity for regulation by cholesterol. Meanwhile, gain-of-Src binding mutations in α2 partially restored the regulation. Furthermore, through studies in caveolin-1 knockdown cells, as well as subcellular distribution studies in cell lines with different α-isoforms, we found that Na/K-ATPase, Src, and caveolin-1 worked together for the cholesterol regulation. Taken together, these new findings reveal that the putative Src-binding domain and the intact Na/K-ATPase/Src/caveolin-1 complex are indispensable for the isoform-specific regulation of Na/K-ATPase by cholesterol.
Collapse
Affiliation(s)
- Jue Zhang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia.,Blood Research Institute, Versiti, Milwaukee, Wisconsin
| | - Xin Li
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Yu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Isabel Larre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Prabhatchandra R Dube
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - David J Kennedy
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - W H Wilson Tang
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Kristen Westfall
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, 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
| | - Yiliang Chen
- Blood Research Institute, Versiti, Milwaukee, Wisconsin.,Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
9
|
Alphavirus Replication: The Role of Cardiac Glycosides and Ion Concentration in Host Cells. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2813253. [PMID: 32461975 PMCID: PMC7232666 DOI: 10.1155/2020/2813253] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/31/2020] [Accepted: 04/15/2020] [Indexed: 01/07/2023]
Abstract
Alphaviruses are arthropod-borne viruses that can cause fever, rash, arthralgias, and encephalitis. The mosquito species Aedes aegypti and Aedes albopictus are the most frequent transmitters of alphaviruses. There are no effective vaccines or specific antivirals available for the treatment of alphavirus-related infections. Interestingly, changes in ion concentration in host cells have been characterized as critical regulators of the alphavirus life cycle, including fusion with the host cell, glycoprotein trafficking, genome translation, and viral budding. Cardiac glycosides, which are classical inhibitors of the Na+ K+ ATPase (NKA), can inhibit alphavirus replication although their mechanisms of action are poorly understood. Nonetheless, results from multiple studies suggest that inhibition of NKA may be a suitable strategy for the development of alphavirus-specific antiviral treatments. This review is aimed at exploring the role of changes in ion concentration during alphavirus replication and at considering the possibility of NKA as a potential therapeutic target for antiviral drugs.
Collapse
|
10
|
Liu J, Nie Y, Chaudhry M, Bai F, Chuang J, Sodhi K, Shapiro JI. The Redox-Sensitive Na/K-ATPase Signaling in Uremic Cardiomyopathy. Int J Mol Sci 2020; 21:ijms21041256. [PMID: 32069992 PMCID: PMC7072896 DOI: 10.3390/ijms21041256] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 02/07/2023] Open
Abstract
In recent years, Na/K-ATPase signaling has been implicated in different physiological and pathophysiological conditions, including cardiac hypertrophy and uremic cardiomyopathy. Cardiotonic steroids (CTS), specific ligands of Na/K-ATPase, regulate its enzymatic activity (at higher concentrations) and signaling function (at lower concentrations without significantly affecting its enzymatic activity) and increase reactive oxygen species (ROS) generation. On the other hand, an increase in ROS alone also regulates the Na/K-ATPase enzymatic activity and signaling function. We termed this phenomenon the Na/K-ATPase-mediated oxidant-amplification loop, in which oxidative stress regulates both the Na/K-ATPase activity and signaling. Most recently, we also demonstrated that this amplification loop is involved in the development of uremic cardiomyopathy. This review aims to evaluate the redox-sensitive Na/K-ATPase-mediated oxidant amplification loop and uremic cardiomyopathy.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (Y.N.); (M.C.); (F.B.)
- Correspondence:
| | - Ying Nie
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (Y.N.); (M.C.); (F.B.)
| | - Muhammad Chaudhry
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (Y.N.); (M.C.); (F.B.)
| | - Fang Bai
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (Y.N.); (M.C.); (F.B.)
| | - Justin Chuang
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (J.C.); (K.S.); (J.I.S.)
| | - Komal Sodhi
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (J.C.); (K.S.); (J.I.S.)
| | - Joseph I. Shapiro
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (J.C.); (K.S.); (J.I.S.)
| |
Collapse
|
11
|
Blaustein MP, Hamlyn JM. Ouabain, endogenous ouabain and ouabain-like factors: The Na + pump/ouabain receptor, its linkage to NCX, and its myriad functions. Cell Calcium 2020; 86:102159. [PMID: 31986323 DOI: 10.1016/j.ceca.2020.102159] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/01/2020] [Accepted: 01/03/2020] [Indexed: 12/12/2022]
Abstract
In this brief review we discuss some aspects of the Na+ pump and its roles in mediating the effects of ouabain and endogenous ouabain (EO): i) in regulating the cytosolic Ca2+ concentration ([Ca2+]CYT) via Na/Ca exchange (NCX), and ii) in activating a number of protein kinase (PK) signaling cascades that control a myriad of cell functions. Importantly, [Ca2+]CYT and the other signaling pathways intersect at numerous points because of the influence of Ca2+ and calmodulin in modulating some steps in those other pathways. While both mechanisms operate in virtually all cells and tissues, this article focuses primarily on their functions in the cardiovascular system, the central nervous system (CNS) and the kidneys.
Collapse
Affiliation(s)
- Mordecai P Blaustein
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - John M Hamlyn
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
12
|
Zhao GH, Qiu YQ, Yang CW, Chen IS, Chen CY, Lee SJ. The cardenolides ouabain and reevesioside A promote FGF2 secretion and subsequent FGFR1 phosphorylation via converged ERK1/2 activation. Biochem Pharmacol 2019; 172:113741. [PMID: 31812679 DOI: 10.1016/j.bcp.2019.113741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 12/02/2019] [Indexed: 11/16/2022]
Abstract
Na+/K+-ATPase α1 was reported to directly interact with and recruit FGF2 (fibroblast growth factor 2), a vital cell signaling protein implicated in angiogenesis, to the inner plasma membrane for subsequent secretion. Cardenolides, a class of cardiac glycosides, were reported to downregulate FGF2 secretion upon binding to Na+/K+-ATPase α1 in a cell system with ectopically expressed FGF2 and Na+/K+-ATPase α1. Herein, we disclose that the cardenolides ouabain and reevesioside A significantly enhance the secretion/release of FGF2 and the phosphorylation of FGFR1 (fibroblast growth factor receptor 1) in a time- and dose-dependent manner, in A549 carcinoma cells. A pharmacological approach was used to elucidate the pertinent upstream effectors. Only the ERK1/2 inhibitor U0126 but not the other inhibitors examined (including those inhibiting the unconventional secretion of FGF2) was able to reduce ouabain-induced FGF2 secretion and FGFR1 activation. ERK1/2 phosphorylation was increased upon ouabain treatment, a process found to be mediated through upstream effectors including ouabain-induced phosphorylated EGFR and a reduced MKP1 protein level. Therefore, at least two independent lines of upstream effectors are able to mediate ouabain-induced ERK1/2 phosphorylation and the subsequent FGF2 secretion and FGFR1 activation. These finding constitute unprecedent insights into the regulation of FGF2 secretion by cardenolides.
Collapse
Affiliation(s)
- Guan-Hao Zhao
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan, ROC; Department of Life Sciences, National Central University, Taoyuan 32001, Taiwan, ROC
| | - Ya-Qi Qiu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan, ROC
| | - Cheng-Wei Yang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan, ROC
| | - Ih-Sheng Chen
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Chin-Yu Chen
- Department of Life Sciences, National Central University, Taoyuan 32001, Taiwan, ROC
| | - Shiow-Ju Lee
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan, ROC.
| |
Collapse
|
13
|
Telocinobufagin and Marinobufagin Produce Different Effects in LLC-PK1 Cells: A Case of Functional Selectivity of Bufadienolides. Int J Mol Sci 2018; 19:ijms19092769. [PMID: 30223494 PMCID: PMC6163863 DOI: 10.3390/ijms19092769] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/09/2018] [Accepted: 09/13/2018] [Indexed: 12/24/2022] Open
Abstract
Bufadienolides are cardiotonic steroids (CTS) identified in mammals. Besides Na+/K+-ATPase inhibition, they activate signal transduction via protein–protein interactions. Diversity of endogenous bufadienolides and mechanisms of action may indicate the presence of functional selectivity and unique cellular outcomes. We evaluated whether the bufadienolides telocinobufagin and marinobufagin induce changes in proliferation or viability of pig kidney (LLC-PK1) cells and the mechanisms involved in these changes. In some experiments, ouabain was used as a positive control. CTS exhibited an inhibitory IC50 of 0.20 (telocinobufagin), 0.14 (ouabain), and 3.40 μM (marinobufagin) for pig kidney Na+/K+-ATPase activity and concentrations that barely inhibited it were tested in LLC-PK1 cells. CTS induced rapid ERK1/2 phosphorylation, but corresponding proliferative response was observed for marinobufagin and ouabain instead of telocinobufagin. Telocinobufagin increased Bax:Bcl-2 expression ratio, sub-G0 cell cycle phase and pyknotic nuclei, indicating apoptosis. Src and MEK1/2 inhibitors blunted marinobufagin but not telocinobufagin effect, which was also not mediated by p38, JNK1/2, and PI3K. However, BIO, a GSK-3β inhibitor, reduced proliferation and, as telocinobufagin, phosphorylated GSK-3β at inhibitory Ser9. Combination of both drugs resulted in synergistic antiproliferative effect. Wnt reporter activity assay showed that telocinobufagin impaired Wnt/β-catenin pathway by acting upstream to β-catenin stabilization. Our findings support that mammalian endogenous bufadienolides may exhibit functional selectivity.
Collapse
|
14
|
Abstract
Caveolin-1 (Cav1) is essential for the formation of caveolae. Little is known about their functional role in the kidney. We tested the hypothesis that caveolae modulate renal salt and water reabsorption. Wild-type (WT) and Cav1-deficient (Cav1−/−) mice were studied. Cav1 expression and caveolae formation were present in vascular cells, late distal convoluted tubule and principal connecting tubule and collecting duct cells of WT but not Cav1−/− kidneys. Urinary sodium excretion was increased by 94% and urine flow by 126% in Cav1−/− mice (p < 0.05). A decrease in activating phosphorylation of the Na-Cl cotransporter (NCC) of the distal convoluted tubule was recorded in Cav1−/− compared to WT kidneys (−40%; p < 0.05). Isolated intrarenal arteries from Cav1−/− mice revealed a fourfold reduction in sensitivity to phenylephrine (p < 0.05). A significantly diminished maximal contractile response (−13%; p < 0.05) was suggestive of enhanced nitric oxide (NO) availability. In line with this, the abundance of endothelial NO synthase (eNOS) was increased in Cav1−/− kidneys +213%; p < 0.05) and cultured caveolae-deprived cells showed intracellular accumulation of eNOS, compared to caveolae-intact controls. Our results suggest that renal caveolae help to conserve water and electrolytes via modulation of NCC function and regulation of vascular eNOS.
Collapse
|
15
|
Pereira DG, Salgado MA, Rocha SC, Santos HL, Villar JA, Contreras RG, Fontes CF, Barbosa LA, Cortes VF. Involvement of Src signaling in the synergistic effect between cisplatin and digoxin on cancer cell viability. J Cell Biochem 2017; 119:3352-3362. [DOI: 10.1002/jcb.26499] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 11/09/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Duane G. Pereira
- Faculdade de Bioquimica, Laboratorio de Bioquimica Celular, Universidade Federal de São João Del ReiCampus Centro‐Oeste Dona LinduDivinopolis, MGBrazil
| | - Mariana A.R. Salgado
- Faculdade de Bioquimica, Laboratorio de Bioquimica Celular, Universidade Federal de São João Del ReiCampus Centro‐Oeste Dona LinduDivinopolis, MGBrazil
| | - Sayonarah C. Rocha
- Faculdade de Bioquimica, Laboratorio de Bioquimica Celular, Universidade Federal de São João Del ReiCampus Centro‐Oeste Dona LinduDivinopolis, MGBrazil
| | - Hérica L. Santos
- Faculdade de Bioquimica, Laboratorio de Bioquimica Celular, Universidade Federal de São João Del ReiCampus Centro‐Oeste Dona LinduDivinopolis, MGBrazil
| | - José A.F.P. Villar
- Laboratório de Síntese Orgânica e Nanoestruturas, Universidade Federal de São João del ReiCampus Centro‐Oeste Dona LinduDivinopolis, MGBrazil
| | - Rubén G. Contreras
- Department of Physiology, Biophysics and NeurosciencesCenter for Research and Advanced Studies (Cinvestav)Mexico CityMexico
| | - Carlos F.L. Fontes
- Laboratório de Estrutura e Regulação de Proteínas e ATPases, Instituto de Bioquimica Médica Leopoldo de Meis, Centro de Ciências da SaúdeUniversidade Federal do Rio de JaneiroRio de Janeiro, RJBrazil
| | - Leandro A. Barbosa
- Faculdade de Bioquimica, Laboratorio de Bioquimica Celular, Universidade Federal de São João Del ReiCampus Centro‐Oeste Dona LinduDivinopolis, MGBrazil
| | - Vanessa F. Cortes
- Faculdade de Bioquimica, Laboratorio de Bioquimica Celular, Universidade Federal de São João Del ReiCampus Centro‐Oeste Dona LinduDivinopolis, MGBrazil
| |
Collapse
|
16
|
Blaustein MP. The pump, the exchanger, and the holy spirit: origins and 40-year evolution of ideas about the ouabain-Na + pump endocrine system. Am J Physiol Cell Physiol 2017; 314:C3-C26. [PMID: 28971835 DOI: 10.1152/ajpcell.00196.2017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Two prescient 1953 publications set the stage for the elucidation of a novel endocrine system: Schatzmann's report that cardiotonic steroids (CTSs) are all Na+ pump inhibitors, and Szent-Gyorgi's suggestion that there is an endogenous "missing screw" in heart failure that CTSs like digoxin may replace. In 1977 I postulated that an endogenous Na+ pump inhibitor acts as a natriuretic hormone and simultaneously elevates blood pressure (BP) in salt-dependent hypertension. This hypothesis was based on the idea that excess renal salt retention promoted the secretion of a CTS-like hormone that inhibits renal Na+ pumps and salt reabsorption. The hormone also inhibits arterial Na+ pumps, elevates myocyte Na+ and promotes Na/Ca exchanger-mediated Ca2+ gain. This enhances vasoconstriction and arterial tone-the hallmark of hypertension. Here I describe how those ideas led to the discovery that the CTS-like hormone is endogenous ouabain (EO), a key factor in the pathogenesis of hypertension and heart failure. Seminal observations that underlie the still-emerging picture of the EO-Na+ pump endocrine system in the physiology and pathophysiology of multiple organ systems are summarized. Milestones include: 1) cloning the Na+ pump isoforms and physiological studies of mutated pumps in mice; 2) discovery that Na+ pumps are also EO-triggered signaling molecules; 3) demonstration that ouabain, but not digoxin, is hypertensinogenic; 4) elucidation of EO's roles in kidney development and cardiovascular and renal physiology and pathophysiology; 5) discovery of "brain ouabain", a component of a novel hypothalamic neuromodulatory pathway; and 6) finding that EO and its brain receptors modulate behavior and learning.
Collapse
Affiliation(s)
- Mordecai P Blaustein
- Departments of Physiology and Medicine, University of Maryland School of Medicine , Baltimore, Maryland
| |
Collapse
|
17
|
Plössl K, Royer M, Bernklau S, Tavraz NN, Friedrich T, Wild J, Weber BHF, Friedrich U. Retinoschisin is linked to retinal Na/K-ATPase signaling and localization. Mol Biol Cell 2017; 28:2178-2189. [PMID: 28615319 PMCID: PMC5531734 DOI: 10.1091/mbc.e17-01-0064] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 01/30/2023] Open
Abstract
Retinoschisin binds to the extracellular domain of Na/K-ATPase subunit β2. Retinoschisin inhibits Na/K-ATPase–associated signaling cascades and affects Na/K-ATPase localization. The retinoschisin-Na/K-ATPase complex overlaps with signaling mediators. Defective Na/K-ATPase signaling by retinoschisin deficiency may promote retinal dystrophy. Mutations in the RS1 gene cause X-linked juvenile retinoschisis (XLRS), a hereditary retinal dystrophy. We recently showed that retinoschisin, the protein encoded by RS1, regulates ERK signaling and apoptosis in retinal cells. In this study, we explored an influence of retinoschisin on the functionality of the Na/K-ATPase, its interaction partner at retinal plasma membranes. We show that retinoschisin binding requires the β2-subunit of the Na/K-ATPase, whereas the α-subunit is exchangeable. Our investigations revealed no effect of retinoschisin on Na/K-ATPase–mediated ATP hydrolysis and ion transport. However, we identified an influence of retinoschisin on Na/K-ATPase–regulated signaling cascades and Na/K-ATPase localization. In addition to the known ERK deactivation, retinoschisin treatment of retinoschisin-deficient (Rs1h-/Y) murine retinal explants decreased activation of Src, an initial transmitter in Na/K-ATPase signal transduction, and of Ca2+ signaling marker Camk2. Immunohistochemistry on murine retinae revealed an overlap of the retinoschisin–Na/K-ATPase complex with proteins involved in Na/K-ATPase signaling, such as caveolin, phospholipase C, Src, and the IP3 receptor. Finally, retinoschisin treatment altered Na/K-ATPase localization in photoreceptors of Rs1h-/Y retinae. Taken together, our results suggest a regulatory effect of retinoschisin on Na/K-ATPase signaling and localization, whereas Na/K-ATPase-dysregulation caused by retinoschisin deficiency could represent an initial step in XLRS pathogenesis.
Collapse
Affiliation(s)
- Karolina Plössl
- Institute of Human Genetics, University of Regensburg, 93053 Regensburg, Germany
| | - Melanie Royer
- Institute of Human Genetics, University of Regensburg, 93053 Regensburg, Germany
| | - Sarah Bernklau
- Institute of Human Genetics, University of Regensburg, 93053 Regensburg, Germany
| | - Neslihan N Tavraz
- Institute of Chemistry, Technical University of Berlin, 10623 Berlin, Germany
| | - Thomas Friedrich
- Institute of Chemistry, Technical University of Berlin, 10623 Berlin, Germany
| | - Jens Wild
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, 93053 Regensburg, Germany
| | - Bernhard H F Weber
- Institute of Human Genetics, University of Regensburg, 93053 Regensburg, Germany
| | - Ulrike Friedrich
- Institute of Human Genetics, University of Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
18
|
Cui X, Xie Z. Protein Interaction and Na/K-ATPase-Mediated Signal Transduction. Molecules 2017; 22:molecules22060990. [PMID: 28613263 PMCID: PMC6152704 DOI: 10.3390/molecules22060990] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/01/2017] [Accepted: 06/02/2017] [Indexed: 02/05/2023] Open
Abstract
The Na/K-ATPase (NKA), or Na pump, is a member of the P-type ATPase superfamily. In addition to pumping ions across cell membrane, it is engaged in assembly of multiple protein complexes in the plasma membrane. This assembly allows NKA to perform many non-pumping functions including signal transduction that are important for animal physiology and disease progression. This article will focus on the role of protein interaction in NKA-mediated signal transduction, and its potential utility as target for developing new therapeutics.
Collapse
Affiliation(s)
- Xiaoyu Cui
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA.
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA.
| |
Collapse
|
19
|
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: 13] [Impact Index Per Article: 1.6] [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
|
20
|
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
|
21
|
Na/K Pump and Beyond: Na/K-ATPase as a Modulator of Apoptosis and Autophagy. Molecules 2017; 22:molecules22040578. [PMID: 28430151 PMCID: PMC6154632 DOI: 10.3390/molecules22040578] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/29/2017] [Accepted: 03/29/2017] [Indexed: 01/22/2023] Open
Abstract
Lung cancer is a leading cause of global cancer deaths. Na/K-ATPase has been studied as a target for cancer treatment. Cardiotonic steroids (CS) trigger intracellular signalling upon binding to Na/K-ATPase. Normal lung and tumour cells frequently express different pump isoforms. Thus, Na/K-ATPase is a powerful target for lung cancer treatment. Drugs targeting Na/K-ATPase may induce apoptosis and autophagy in transformed cells. We argue that Na/K-ATPase has a role as a potential target in chemotherapy in lung cancer treatment. We discuss the effects of Na/K-ATPase ligands and molecular pathways inducing deleterious effects on lung cancer cells, especially those leading to apoptosis and autophagy.
Collapse
|
22
|
Zhou HT, Yu XF, Zhou GM. Diosgenin inhibits angiotensin II-induced extracellular matrix remodeling in cardiac fibroblasts through regulating the TGF-β1/Smad3 signaling pathway. Mol Med Rep 2017; 15:2823-2828. [DOI: 10.3892/mmr.2017.6280] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 11/15/2016] [Indexed: 11/05/2022] Open
|
23
|
Madan N, Xu Y, Duan Q, Banerjee M, Larre I, Pierre SV, Xie Z. Src-independent ERK signaling through the rat α3 isoform of Na/K-ATPase. Am J Physiol Cell Physiol 2017; 312:C222-C232. [PMID: 27903584 PMCID: PMC5401946 DOI: 10.1152/ajpcell.00199.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 11/30/2016] [Accepted: 11/30/2016] [Indexed: 01/10/2023]
Abstract
The Na/K-ATPase α1 polypeptide supports both ion-pumping and signaling functions. The Na/K-ATPase α3 polypeptide differs from α1 in both its primary structure and its tissue distribution. The expression of α3 seems particularly important in neurons, and recent clinical evidence supports a unique role of this isoform in normal brain function. The nature of this specific role of α3 has remained elusive, because the ubiquitous presence of α1 has hindered efforts to characterize α3-specific functions in mammalian cell systems. Using Na/K-ATPase α1 knockdown pig kidney cells (PY-17), we generated the first stable mammalian cell line expressing a ouabain-resistant form of rat Na/K-ATPase α3 in the absence of endogenous pig α1 detectable by Western blotting. In these cells, Na/K-ATPase α3 formed a functional ion-pumping enzyme and rescued the expression of Na/K-ATPase β1 and caveolin-1 to levels comparable with those observed in PY-17 cells rescued with a rat Na/K-ATPase α1 (AAC-19). The α3-containing enzymes had lower Na+ affinity and lower ouabain-sensitive transport activity than their α1-containing counterparts under basal conditions, but showed a greater capacity to be activated when intracellular Na+ was increased. In contrast to Na/K-ATPase α1, α3 could not regulate Src. Upon exposure to ouabain, Src activation did not occur, yet ERK was activated through Src-independent pathways involving PI3K and PKC. Hence, α3 expression confers signaling and pumping properties that are clearly distinct from that of cells expressing Na/K-ATPase α1.
Collapse
Affiliation(s)
- Namrata Madan
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Yunhui Xu
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China; and
| | - Qiming Duan
- Gladstone Institute of Cardiovascular Disease, San Francisco, California
| | - Moumita Banerjee
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - Isabel Larre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia
| | - 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
|
24
|
La J, Reed E, Chan L, Smolyaninova LV, Akomova OA, Mutlu GM, Orlov SN, Dulin NO. Downregulation of TGF-β Receptor-2 Expression and Signaling through Inhibition of Na/K-ATPase. PLoS One 2016; 11:e0168363. [PMID: 28006004 PMCID: PMC5179089 DOI: 10.1371/journal.pone.0168363] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Accepted: 11/30/2016] [Indexed: 01/06/2023] Open
Abstract
Transforming growth factor-beta (TGF-β) is a multi-functional cytokine implicated in the control of cell growth and differentiation. TGF-β signals through a complex of TGF-β receptors 1 and 2 (TGFβR1 and TGFβR2) that phosphorylate and activate Smad2/3 transcription factors driving transcription of the Smad-target genes. The Na+/K+-ATPase is an integral plasma membrane protein critical for maintaining the electro-chemical gradient of Na+ and K+ in the cell. We found that inhibition of the Na+/K+ ATPase by ouabain results in a dramatic decrease in the expression of TGFβR2 in human lung fibrobalsts (HLF) at the mRNA and protein levels. This was accompanied by inhibition of TGF-β-induced Smad phosphorylation and the expression of TGF-β target genes, such as fibronectin and smooth muscle alpha-actin. Inhibition of Na+/K+ ATPase by an alternative approach (removal of extracellular potassium) had a similar effect in HLF. Finally, treatment of lung alveolar epithelial cells (A549) with ouabain also resulted in the downregulation of TGFβR2, the inhibition of TGF-β-induced Smad phosphorylation and of the expression of mesenchymal markers, vimentin and fibronectin. Together, these data demonstrate a critical role of Na+/K+-ATPase in the control of TGFβR2 expression, TGF-β signaling and cell responses to TGF-β.
Collapse
Affiliation(s)
- Jennifer La
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, IL, United States of America
| | - Eleanor Reed
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, IL, United States of America
| | - Lan Chan
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, IL, United States of America
| | - Larisa V. Smolyaninova
- Laboratory of Biomembranes, Department of Biophysics, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Olga A. Akomova
- Laboratory of Biomembranes, Department of Biophysics, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Gökhan M. Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, IL, United States of America
| | - Sergei N. Orlov
- Laboratory of Biomembranes, Department of Biophysics, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
- Siberian Medical State University, Tomsk, Russia
| | - Nickolai O. Dulin
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, IL, United States of America
- Siberian Medical State University, Tomsk, Russia
| |
Collapse
|
25
|
Liu M, Feng LX, Sun P, Liu W, Mi T, Lei M, Wu W, Jiang B, Yang M, Hu L, Guo DA, Liu X. Knockdown of Apolipoprotein E Enhanced Sensitivity of Hep3B Cells to Cardiac Steroids via Regulating Na+/K+-ATPase Signalosome. Mol Cancer Ther 2016; 15:2955-2965. [PMID: 27507851 DOI: 10.1158/1535-7163.mct-15-0961] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 06/29/2016] [Accepted: 07/02/2016] [Indexed: 11/16/2022]
Abstract
This study compared the sensitivity of human hepatoma Hep3B, SK-HEP-1, SMMC-7721, and BEL-7402 cells to cardiac steroids, including bufalin (BF), a bufalin derivative (BF211), ouabain (OUA), and digitoxin (DIG). Hep3B cells exhibited relatively low sensitivity to cardiac steroids. Expression levels of subunits of Na+/K+-ATPase were high in Hep3B cells. However, colocalization of Na+/K+-ATPase and caveolin was nearly undetectable in Hep3B cells. By using RNA-Seq technology, we found a total of 36 genes to be differentially expressed between Hep3B cells and SK-HEP-1 cells, which are highly sensitive to cardiac steroids. Our bioinformatics analysis determined that these genes were mostly comprised of extracellular space, protein binding, and extracellular region. Among these 36 genes, apolipoprotein E (APOE) played a critical role, as knockdown APOE expression induced colocalization of Na+/K+-ATPase and caveolin and increased sensitivity of Hep3B cells to both proliferation-inhibiting and cytotoxic effects of BF or BF211. Also, the effects of BF on PI3K/AKT/GSK3β and apoptosis signal cascades were enhanced in APOE knockdown cells. The results of our study confirmed the role of Na+/K+-ATPase signalosome in cytotoxicity of cardiac steroids and suggested that APOE regulated the sensitivity of cells to cardiac steroids by affecting formation and function of Na+/K+-ATPase signalosome. In addition, intercellular interaction with high level of Na+/K+-ATPase β1 subunit may be also a factor in the low sensitivity of Hep3B cells to cardiac steroids. Mol Cancer Ther; 15(12); 2955-65. ©2016 AACR.
Collapse
Affiliation(s)
- Miao Liu
- Shanghai Research Center for Modernization of TCM, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Li-Xing Feng
- Shanghai Research Center for Modernization of TCM, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Peng Sun
- Shanghai Research Center for Modernization of TCM, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Wang Liu
- Shanghai Research Center for Modernization of TCM, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Tian Mi
- Shanghai Research Center for Modernization of TCM, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Min Lei
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Wanying Wu
- Shanghai Research Center for Modernization of TCM, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Baohong Jiang
- Shanghai Research Center for Modernization of TCM, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Min Yang
- Shanghai Research Center for Modernization of TCM, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Lihong Hu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China.
| | - De-An Guo
- Shanghai Research Center for Modernization of TCM, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China.
| | - Xuan Liu
- Shanghai Research Center for Modernization of TCM, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China. .,Department of Cardiology, Shanghai Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P.R. China
| |
Collapse
|
26
|
Kinoshita PF, Leite JA, Orellana AMM, Vasconcelos AR, Quintas LEM, Kawamoto EM, Scavone C. The Influence of Na(+), K(+)-ATPase on Glutamate Signaling in Neurodegenerative Diseases and Senescence. Front Physiol 2016; 7:195. [PMID: 27313535 PMCID: PMC4890531 DOI: 10.3389/fphys.2016.00195] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/17/2016] [Indexed: 12/17/2022] Open
Abstract
Decreased Na(+), K(+)-ATPase (NKA) activity causes energy deficiency, which is commonly observed in neurodegenerative diseases. The NKA is constituted of three subunits: α, β, and γ, with four distinct isoforms of the catalytic α subunit (α1-4). Genetic mutations in the ATP1A2 gene and ATP1A3 gene, encoding the α2 and α3 subunit isoforms, respectively can cause distinct neurological disorders, concurrent to impaired NKA activity. Within the central nervous system (CNS), the α2 isoform is expressed mostly in glial cells and the α3 isoform is neuron-specific. Mutations in ATP1A2 gene can result in familial hemiplegic migraine (FHM2), while mutations in the ATP1A3 gene can cause Rapid-onset dystonia-Parkinsonism (RDP) and alternating hemiplegia of childhood (AHC), as well as the cerebellar ataxia, areflexia, pescavus, optic atrophy and sensorineural hearing loss (CAPOS) syndrome. Data indicates that the central glutamatergic system is affected by mutations in the α2 isoform, however further investigations are required to establish a connection to mutations in the α3 isoform, especially given the diagnostic confusion and overlap with glutamate transporter disease. The age-related decline in brain α2∕3 activity may arise from changes in the cyclic guanosine monophosphate (cGMP) and cGMP-dependent protein kinase (PKG) pathway. Glutamate, through nitric oxide synthase (NOS), cGMP and PKG, stimulates brain α2∕3 activity, with the glutamatergic N-methyl-D-aspartate (NMDA) receptor cascade able to drive an adaptive, neuroprotective response to inflammatory and challenging stimuli, including amyloid-β. Here we review the NKA, both as an ion pump as well as a receptor that interacts with NMDA, including the role of NKA subunits mutations. Failure of the NKA-associated adaptive response mechanisms may render neurons more susceptible to degeneration over the course of aging.
Collapse
Affiliation(s)
- Paula F. Kinoshita
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Jacqueline A. Leite
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Ana Maria M. Orellana
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Andrea R. Vasconcelos
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Luis E. M. Quintas
- Laboratory of Biochemical and Molecular Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Elisa M. Kawamoto
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Cristoforo Scavone
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| |
Collapse
|
27
|
Wu D, Lei H, Wang JY, Zhang CL, Feng H, Fu FY, Li L, Wu LL. CTRP3 attenuates post-infarct cardiac fibrosis by targeting Smad3 activation and inhibiting myofibroblast differentiation. J Mol Med (Berl) 2015; 93:1311-25. [PMID: 26138247 DOI: 10.1007/s00109-015-1309-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 05/31/2015] [Accepted: 06/03/2015] [Indexed: 01/19/2023]
Abstract
UNLABELLED C1q/tumor necrosis factor-related protein-3 (CTRP3) is a novel adipokine with modulation effects on metabolism, inflammation, and cardiovascular system. This study aimed to investigate the effect of CTRP3 on cardiac fibrosis and its underlying mechanism. The myocardial expression of CTRP3 was significantly decreased after myocardial infarction (MI). Adenovirus-delivered CTRP3 supplement attenuated myocardial hypertrophy, improved cardiac function, inhibited interstitial fibrosis, and decreased the number of myofibroblasts post-MI. In cultured adult rat cardiac fibroblasts (CFs), CTRP3 attenuated cell proliferation; migration; and the expression of connective tissue growth factor, collagen I, and collagen III induced by transforming growth factor (TGF)-β1. Moreover, CTRP3 inhibited whereas CTRP3 small interfering RNA (siRNA) facilitated the expression of α-SMA and profibrotic molecules induced by TGF-β1. CTRP3 also attenuated TGF-β1-induced Smad3 phosphorylation, nuclear translocation, and interaction with p300. CTRP3 increased the phosphorylation of AMP-activated protein kinase (AMPK) and Akt in both rat hearts and CFs. Adenine 9-β-D-arabinofuranoside (AraA), an AMPK inhibitor, abolished the protective effect of CTRP3 against TGF-β1-induced profibrotic response and Smad3 activation. Taken together, CTRP3 attenuates cardiac fibrosis by inhibiting myofibroblast differentiation and the subsequent extracellular matrix production. AMPK is required for the anti-fibrotic effect of CTRP3 through targeting Smad3 activation and inhibiting myofibroblast differentiation. KEY MESSAGE CTRP3 alleviates cardiac fibrosis in a rat post-MI model and in cardiac fibroblasts. CTRP3 inhibits fibroblast-to-myofibroblast differentiation. CTRP3 exerts anti-fibrotic effect through targeting Smad3 activation. AMPK mediates the anti-fibrotic effect of CTRP3 by inhibition of Smad3 activation.
Collapse
Affiliation(s)
- Dan Wu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Hong Lei
- Department of Physiology and Pathophysiology, Peking University Health Science Center, and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Jin-Yu Wang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Cheng-Lin Zhang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Han Feng
- Department of Physiology and Pathophysiology, Peking University Health Science Center, and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Feng-Ying Fu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Li Li
- Department of Physiology and Pathophysiology, Peking University Health Science Center, and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| | - Li-Ling Wu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| |
Collapse
|
28
|
Oliveira TN, Possidonio AC, Soares CP, Ayres R, Costa ML, Quintas LEM, Mermelstein C. The role of Na+/K+-ATPase during chick skeletal myogenesis. PLoS One 2015; 10:e0120940. [PMID: 25775465 PMCID: PMC4361648 DOI: 10.1371/journal.pone.0120940] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 02/09/2015] [Indexed: 12/14/2022] Open
Abstract
The formation of a vertebrate skeletal muscle fiber involves a series of sequential and interdependent events that occurs during embryogenesis. One of these events is myoblast fusion which has been widely studied, yet not completely understood. It was previously shown that during myoblast fusion there is an increase in the expression of Na+/K+-ATPase. This fact prompted us to search for a role of the enzyme during chick in vitro skeletal myogenesis. Chick myogenic cells were treated with the Na+/K+-ATPase inhibitor ouabain in four different concentrations (0.01-10 μM) and analyzed. Our results show that 0.01, 0.1 and 1 μM ouabain did not induce changes in cell viability, whereas 10 μM induced a 45% decrease. We also observed a reduction in the number and thickness of multinucleated myotubes and a decrease in the number of myoblasts after 10 μM ouabain treatment. We tested the involvement of MEK-ERK and p38 signaling pathways in the ouabain-induced effects during myogenesis, since both pathways have been associated with Na+/K+-ATPase. The MEK-ERK inhibitor U0126 alone did not alter cell viability and did not change ouabain effect. The p38 inhibitor SB202190 alone or together with 10 μM ouabain did not alter cell viability. Our results show that the 10 μM ouabain effects in myofiber formation do not involve the MEK-ERK or the p38 signaling pathways, and therefore are probably related to the pump activity function of the Na+/K+-ATPase.
Collapse
Affiliation(s)
- Taissa Neustadt Oliveira
- Laboratório de Diferenciação Muscular e Citoesqueleto, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Claudia Possidonio
- Laboratório de Diferenciação Muscular e Citoesqueleto, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carolina Pontes Soares
- Laboratório de Diferenciação Muscular e Citoesqueleto, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo Ayres
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Manoel Luis Costa
- Laboratório de Diferenciação Muscular e Citoesqueleto, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luis Eduardo Menezes Quintas
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cláudia Mermelstein
- Laboratório de Diferenciação Muscular e Citoesqueleto, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
29
|
Lan JY, Williams C, Levin M, Black LD. Depolarization of Cellular Resting Membrane Potential Promotes Neonatal Cardiomyocyte Proliferation In Vitro. Cell Mol Bioeng 2014; 7:432-445. [PMID: 25295125 DOI: 10.1007/s12195-014-0346-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Cardiomyocytes (CMs) undergo a rapid transition from hyperplastic to hypertrophic growth soon after birth, which is a major challenge to the development of engineered cardiac tissue for pediatric patients. Resting membrane potential (Vmem) has been shown to play an important role in cell differentiation and proliferation during development. We hypothesized that depolarization of neonatal CMs would stimulate or maintain CM proliferation in vitro. To test our hypothesis, we isolated postnatal day 3 neonatal rat CMs and subjected them to sustained depolarization via the addition of potassium gluconate or Ouabain to the culture medium. Cell density and CM percentage measurements demonstrated an increase in mitotic CMs along with a ~2 fold increase in CM numbers with depolarization. In addition, depolarization led to an increase in cells in G2 and S phase, indicating increased proliferation, as measured by flow cytometry. Surprisingly depolarization of Vmem with either treatment led to inhibition of proliferation in cardiac fibroblasts. This effect is abrogated when the study was carried out on postnatal day 7 neonatal CMs, which are less proliferative, indicating that the likely mechanism of depolarization is the maintenance of the proliferating CM population. In summary, our findings suggest that depolarization maintains postnatal CM proliferation and may be a novel approach to encourage growth of engineered tissue and cardiac regeneration in pediatric patients.
Collapse
Affiliation(s)
- Jen-Yu Lan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155 USA
| | - Corin Williams
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155 USA
| | - Michael Levin
- Department of Biology, Tufts University, Medford, MA 02155 USA ; Center for Regenerative and Developmental Biology, Tufts University, Medford, MA 02155 USA
| | - Lauren Deems Black
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155 USA ; Cellular, Molecular and Developmental Biology Program, Sackler Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111 USA
| |
Collapse
|
30
|
Affiliation(s)
- Mordecai P Blaustein
- Departments of Physiology and Medicine and the Center for Heart, Hypertension and Kidney Disease, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
31
|
Khundmiri SJ. Advances in understanding the role of cardiac glycosides in control of sodium transport in renal tubules. J Endocrinol 2014; 222:R11-24. [PMID: 24781255 DOI: 10.1530/joe-13-0613] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cardiotonic steroids have been used for the past 200 years in the treatment of congestive heart failure. As specific inhibitors of membrane-bound Na(+)/K(+) ATPase, they enhance cardiac contractility through increasing myocardial cell calcium concentration in response to the resulting increase in intracellular Na concentration. The half-minimal concentrations of cardiotonic steroids required to inhibit Na(+)/K(+) ATPase range from nanomolar to micromolar concentrations. In contrast, the circulating levels of cardiotonic steroids under physiological conditions are in the low picomolar concentration range in healthy subjects, increasing to high picomolar levels under pathophysiological conditions including chronic kidney disease and heart failure. Little is known about the physiological function of low picomolar concentrations of cardiotonic steroids. Recent studies have indicated that physiological concentrations of cardiotonic steroids acutely stimulate the activity of Na(+)/K(+) ATPase and activate an intracellular signaling pathway that regulates a variety of intracellular functions including cell growth and hypertrophy. The effects of circulating cardiotonic steroids on renal salt handling and total body sodium homeostasis are unknown. This review will focus on the role of low picomolar concentrations of cardiotonic steroids in renal Na(+)/K(+) ATPase activity, cell signaling, and blood pressure regulation.
Collapse
Affiliation(s)
- Syed Jalal Khundmiri
- Division of Nephrology and HypertensionDepartment of MedicineDepartment of Physiology and BiophysicsUniversity of Louisville, 570 S. Preston Street, Louisville, Kentucky 40202, USADivision of Nephrology and HypertensionDepartment of MedicineDepartment of Physiology and BiophysicsUniversity of Louisville, 570 S. Preston Street, Louisville, Kentucky 40202, USA
| |
Collapse
|
32
|
Zhang WZ. An association of metabolic syndrome constellation with cellular membrane caveolae. PATHOBIOLOGY OF AGING & AGE RELATED DISEASES 2014; 4:23866. [PMID: 24563731 PMCID: PMC3926988 DOI: 10.3402/pba.v4.23866] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 01/21/2014] [Accepted: 01/21/2014] [Indexed: 01/19/2023]
Abstract
Metabolic syndrome (MetS) is a cluster of metabolic abnormalities that can predispose an individual to a greater risk of developing type-2 diabetes and cardiovascular diseases. The cluster includes abdominal obesity, dyslipidemia, hypertension, and hyperglycemia - all of which are risk factors to public health. While searching for a link among the aforementioned malaises, clues have been focused on the cell membrane domain caveolae, wherein the MetS-associated active molecules are colocalized and interacted with to carry out designated biological activities. Caveola disarray could induce all of those individual metabolic abnormalities to be present in animal models and humans, providing a new target for therapeutic strategy in the management of MetS.
Collapse
Affiliation(s)
- Wei-Zheng Zhang
- CMP Laboratory, Port Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
33
|
Xie JX, Li X, Xie Z. Regulation of renal function and structure by the signaling Na/K-ATPase. IUBMB Life 2013; 65:991-8. [PMID: 24323927 PMCID: PMC5375025 DOI: 10.1002/iub.1229] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 11/08/2013] [Indexed: 12/23/2022]
Abstract
The Na/K-ATPase as an essential ion pump was discovered more than 50 years ago (Skou (1989) Biochim. Biophys. Acta 1000, 439-446; Feraille and Doucet (2001) Physiol. Rev. 81, 345-418). The signaling function of Na/K-ATPase has been gradually appreciated over the last 20 years, first from the studies of regulatory effects of ouabain on cardiac cell growth. Several reviews on this topic have been written during the last few years (Schoner and Scheiner-Bobis (2007) Am. J. Physiol. Cell. Physiol. 293, C509-C536; Xie and Cai (2003) Mol. Interv. 3, 157 - 168; Bagrov et al. (2009) Pharmacol. Rev. 61, 9-38; Tian and Xie (2008) Physiology 23, 205-211; Fontana et al. (2013) FEBS J. 280, 5450-5455; Blanco and Wallace (2013) Am. J. Physiol. Renal Physiol. 305, F797-F812). This article will focus on the molecular mechanism of Na/K-ATPase-mediated signal transduction and its potential regulatory role in renal physiology and diseases.
Collapse
Affiliation(s)
- Jeffrey X Xie
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, USA
| | | | | |
Collapse
|
34
|
Wu J, Akkuratov EE, Bai Y, Gaskill CM, Askari A, Liu L. Cell signaling associated with Na(+)/K(+)-ATPase: activation of phosphatidylinositide 3-kinase IA/Akt by ouabain is independent of Src. Biochemistry 2013; 52:9059-67. [PMID: 24266852 PMCID: PMC3868411 DOI: 10.1021/bi4011804] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Exposure
of intact cells to selective inhibitors of Na+/K+-ATPase such as ouabain activates several growth-related
cell signaling pathways. It has been suggested that the initial event
of these pathways is the binding of ouabain to a preexisting complex
of Src with Na+/K+-ATPase of the plasma membrane.
The aim of this work was to evaluate the role of Src in the ouabain-induced
activation of phosphatidylinositide 3-kinase 1A (PI3K1A) and its downstream
consequences. When fibroblasts devoid of Src (SYF cells) and controls
(Src++ cells) were exposed to ouabain, PI3K1A, Akt, and
proliferative growth were similarly stimulated in both cell lines.
Ouabain-induced activation of Akt was not prevented by the Src inhibitor
PP2. In contrast, ERK1/2 were not activated by ouabain in SYF cells
but were stimulated in Src++ cells; this was prevented
by PP2. In isolated adult mouse cardiac myocytes, where ouabain induces
hypertrophic growth, PP2 also did not prevent ouabain-induced activation
of Akt and the resulting hypertrophy. Ouabain-induced increases in
the levels of co-immunoprecipitation of the α-subunit of Na+/K+-ATPase with the p85 subunit of PI3K1A were
noted in SYF cells, Src++ cells, and adult cardiac myocytes.
In conjunction with previous findings, the results presented here
indicate that (a) if there is a preformed complex of Src and Na+/K+-ATPase, it is irrelevant to ouabain-induced
activation of the PI3K1A/Akt pathway through Na+/K+-ATPase and (b) a more likely, but not established, mechanism
of linkage of Na+/K+-ATPase to PI3K1A is the
ouabain-induced interaction of a proline-rich domain of the α-subunit
of Na+/K+-ATPase with the SH3 domain of the
p85 subunit of PI3K1A.
Collapse
Affiliation(s)
- Jian Wu
- Department of Biochemistry and Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus , Toledo, Ohio 43614, United States
| | | | | | | | | | | |
Collapse
|
35
|
Blanco G, Wallace DP. Novel role of ouabain as a cystogenic factor in autosomal dominant polycystic kidney disease. Am J Physiol Renal Physiol 2013; 305:F797-812. [PMID: 23761677 DOI: 10.1152/ajprenal.00248.2013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The classic role of the Na-K-ATPase is that of a primary active transporter that utilizes cell energy to establish and maintain transmembrane Na(+) and K(+) gradients to preserve cell osmotic stability, support cell excitability, and drive secondary active transport. Recent studies have revealed that Na-K-ATPase located within cholesterol-containing lipid rafts serves as a receptor for cardiotonic steroids, including ouabain. Traditionally, ouabain was viewed as a toxin produced only in plants, and it was used in relatively high concentrations to experimentally block the pumping action of the Na-K-ATPase. However, the new and unexpected role of the Na-K-ATPase as a signal transducer revealed a novel facet for ouabain in the regulation of a myriad of cell functions, including cell proliferation, hypertrophy, apoptosis, mobility, and metabolism. The seminal discovery that ouabain is endogenously produced in mammals and circulates in plasma has fueled the interest in this endogenous molecule as a potentially important hormone in normal physiology and disease. In this article, we review the role of the Na-K-ATPase as an ion transporter in the kidney, the experimental evidence for ouabain as a circulating hormone, the function of the Na-K-ATPase as a signal transducer that mediates ouabain's effects, and novel results for ouabain-induced Na-K-ATPase signaling in cystogenesis of autosomal dominant polycystic kidney disease.
Collapse
Affiliation(s)
- Gustavo Blanco
- Dept. of Molecular and Integrative Physiology, 3901 Rainbow Blvd., Kansas City, KS 66160.
| | | |
Collapse
|
36
|
Lucas TFG, Amaral LS, Porto CS, Quintas LEM. Na+/K+-ATPase α1 isoform mediates ouabain-induced expression of cyclin D1 and proliferation of rat sertoli cells. Reproduction 2012; 144:737-45. [PMID: 23028124 DOI: 10.1530/rep-12-0232] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Novel roles for the interaction of cardiotonic steroids to Na(+)/K(+)-ATPase have been established in recent years. The aim of this study was to investigate the intracellular signaling events downstream the action of ouabain on Na(+)/K(+)-ATPase in Sertoli cell obtained from immature rats. Treatment of Sertoli cells with ouabain (1 μM) induced a rapid and transient increase in the extracellular signal-regulated kinase (ERK1/2 or MAPK3/1) and phosphatidylinositol 3-kinase (PI3K)/serine-threonine protein kinase (AKT) phosphorylation. Also, ouabain upregulated the expression of cyclin D1 and incorporation of [methyl-(3)H]thymidine, both of which were dependent on MAPK3/1 but not AKT intracellular cascade, as shown by pretreatment with MEK (MAP2K1/2) inhibitor U0126 and PI3K inhibitor wortmannin respectively. Moreover, the effect of ouabain on these proliferation parameters was completely prevented by phospho-cAMP response element-binding protein (CREB)/CREB-binding protein complex inhibitor KG501 and only partially by nuclear factor κB nuclear translocation inhibitor SN50. Pretreatment with estrogen receptor antagonist ICI 182780 showed that MAPK3/1 activation by ouabain does not involve this receptor. The Na(+)/K(+)-ATPase α1 isoform, but not α4, was detected in Sertoli cells, suggesting that ouabain effects in Sertoli cells are mediated via α1. Taken together, these results show a rapid ouabain action in the Sertoli cells, which in turn can modulate nuclear transcriptional events essential for Sertoli cell proliferation in a critical period of testicular development. Our findings are important to understand the role of ouabain in the testis and its possible implications in male infertility.
Collapse
Affiliation(s)
- Thaís F G Lucas
- Setor de Endocrinologia Experimental, Departamento de Farmacologia, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, São Paulo, Brazil
| | | | | | | |
Collapse
|
37
|
Morrill GA, Kostellow AB, Askari A. Caveolin-Na/K-ATPase interactions: role of transmembrane topology in non-genomic steroid signal transduction. Steroids 2012; 77:1160-8. [PMID: 22579740 DOI: 10.1016/j.steroids.2012.04.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 04/04/2012] [Accepted: 04/10/2012] [Indexed: 12/14/2022]
Abstract
Progesterone and its polar metabolite(s) trigger the meiotic divisions in the amphibian oocyte through a non-genomic signaling system at the plasma membrane. Published site-directed mutagenesis studies of ouabain binding and progesterone-ouabain competition studies indicate that progesterone binds to a 23 amino acid extracellular loop of the plasma membrane α-subunit of Na/K-ATPase. Integral membrane proteins such as caveolins are reported to form Na/K-ATPase-peptide complexes essential for signal transduction. We have characterized the progesterone-induced Na/K-ATPase-caveolin (CAV-1)-steroid 5α-reductase interactions initiating the meiotic divisions. Peptide sequence analysis algorithms indicate that CAV-1 contains two plasma membrane spanning helices, separated by as few as 1-2 amino acid residues at the cell surface. The CAV-1 scaffolding domain, reported to interact with CAV-1 binding (CB) motifs in signaling proteins, overlaps transmembrane (TM) helix 1. The α-subunit of Na/K-ATPase (10 TM helices) contains double CB motifs within TM-1 and TM-10. Steroid 5α-reductase (6 TM helices), an initial step in polar steroid formation, contains CB motifs overlapping TM-1 and TM-6. Computer analysis predicts that interaction between antipathic strands may bring CB motifs and scaffolding domains into close proximity, initiating allostearic changes. Progesterone binding to the α-subunit may thus facilitate CB motif:CAV-1 interaction, which in turn induces helix-helix interaction and generates both a signaling cascade and formation of polar steroids.
Collapse
Affiliation(s)
- Gene A Morrill
- Department of Physiology & Biophysics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | | | | |
Collapse
|
38
|
Sjögren B, Parra S, Heath LJ, Atkins KB, Xie ZJ, Neubig RR. Cardiotonic steroids stabilize regulator of G protein signaling 2 protein levels. Mol Pharmacol 2012; 82:500-9. [PMID: 22695717 DOI: 10.1124/mol.112.079293] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Regulator of G protein signaling 2 (RGS2), a G(q)-specific GTPase-activating protein, is strongly implicated in cardiovascular function. RGS2(-/-) mice are hypertensive and prone to heart failure, and several rare human mutations that accelerate RGS2 degradation have been identified among patients with hypertension. Therefore, pharmacological up-regulation of RGS2 protein levels might be beneficial. We used a β-galactosidase complementation method to screen several thousand compounds with known pharmacological functions for those that increased RGS2 protein levels. Several cardiotonic steroids (CTSs), including ouabain and digoxin, increased RGS2 but not RGS4 protein levels. CTSs increased RGS2 protein levels through a post-transcriptional mechanism, by slowing protein degradation. RGS2 mRNA levels in primary vascular smooth muscle cells were unaffected by CTS treatment, whereas protein levels were increased 2- to 3-fold. Na(+)/K(+)-ATPase was required for the increase in RGS2 protein levels, because the effect was lost in Na(+)/K(+)-ATPase-knockdown cells. Furthermore, we demonstrated that CTS-induced increases in RGS2 levels were functional and reduced receptor-stimulated, G(q)-dependent, extracellular signal-regulated kinase phosphorylation. Finally, we showed that in vivo treatment with digoxin led to increased RGS2 protein levels in heart and kidney. CTS-induced increases in RGS2 protein levels and function might modify several deleterious mechanisms in hypertension and heart failure. This novel CTS mechanism might contribute to the beneficial actions of low-dose digoxin treatment in heart failure. Our results support the concept of small-molecule modulation of RGS2 protein levels as a new strategy for cardiovascular therapy.
Collapse
Affiliation(s)
- Benita Sjögren
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|
39
|
Li X, McClellan ME, Tanito M, Garteiser P, Towner R, Bissig D, Berkowitz BA, Fliesler SJ, Woodruff ML, Fain GL, Birch DG, Khan MS, Ash JD, Elliott MH. Loss of caveolin-1 impairs retinal function due to disturbance of subretinal microenvironment. J Biol Chem 2012; 287:16424-34. [PMID: 22451674 DOI: 10.1074/jbc.m112.353763] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Caveolin-1 (Cav-1), an integral component of caveolar membrane domains, is expressed in several retinal cell types, including photoreceptors, retinal vascular endothelial cells, Müller glia, and retinal pigment epithelium (RPE) cells. Recent evidence links Cav-1 to ocular diseases, including autoimmune uveitis, diabetic retinopathy, and primary open angle glaucoma, but its role in normal vision is largely undetermined. In this report, we show that ablation of Cav-1 results in reduced inner and outer retinal function as measured, in vivo, by electroretinography and manganese-enhanced MRI. Somewhat surprisingly, dark current and light sensitivity were normal in individual rods (recorded with suction electrode methods) from Cav-1 knock-out (KO) mice. Although photoreceptor function was largely normal, in vitro, the apparent K(+) affinity of the RPE-expressed α1-Na(+)/K(+)-ATPase was decreased in Cav-1 KO mice. Cav-1 KO retinas also displayed unusually tight adhesion with the RPE, which could be resolved by brief treatment with hyperosmotic medium, suggesting alterations in outer retinal fluid homeostasis. Collectively, these findings demonstrate that reduced retinal function resulting from Cav-1 ablation is not photoreceptor-intrinsic but rather involves impaired subretinal and/or RPE ion/fluid homeostasis.
Collapse
Affiliation(s)
- Xiaoman Li
- Department of Ophthalmology and Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
New Insights into the Regulation of Na+,K+-ATPase by Ouabain. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 294:99-132. [DOI: 10.1016/b978-0-12-394305-7.00002-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
41
|
Increased endothelial cell-leukocyte interaction in murine schistosomiasis: possible priming of endothelial cells by the disease. PLoS One 2011; 6:e23547. [PMID: 21853150 PMCID: PMC3154496 DOI: 10.1371/journal.pone.0023547] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Accepted: 07/19/2011] [Indexed: 11/21/2022] Open
Abstract
Background and Aims Schistosomiasis is an intravascular parasitic disease associated with inflammation. Endothelial cells control leukocyte transmigration and vascular permeability being modulated by pro-inflammatory mediators. Recent data have shown that endothelial cells primed in vivo in the course of a disease keep the information in culture. Herein, we evaluated the impact of schistosomiasis on endothelial cell-regulated events in vivo and in vitro. Methodology and Principal Findings The experimental groups consisted of Schistosoma mansoni-infected and age-matched control mice. In vivo infection caused a marked influx of leukocytes and an increased protein leakage in the peritoneal cavity, characterizing an inflamed vascular and cellular profile. In vitro leukocyte-mesenteric endothelial cell adhesion was higher in cultured cells from infected mice as compared to controls, either in the basal condition or after treatment with the pro-inflammatory cytokine tumor necrosis factor (TNF). Nitric oxide (NO) donation reduced leukocyte adhesion to endothelial cells from control and infected groups; however, in the later group the effect was more pronounced, probably due to a reduced NO production. Inhibition of control endothelial NO synthase (eNOS) increased leukocyte adhesion to a level similar to the one observed in the infected group. Besides, the adhesion of control leukocytes to endothelial cells from infected animals is similar to the result of infected animals, confirming that schistosomiasis alters endothelial cells function. Furthermore, NO production as well as the expression of eNOS were reduced in cultured endothelial cells from infected animals. On the other hand, the expression of its repressor protein, namely caveolin-1, was similar in both control and infected groups. Conclusion/Significance Schistosomiasis increases vascular permeability and endothelial cell-leukocyte interaction in vivo and in vitro. These effects are partially explained by a reduced eNOS expression. In addition, our data show that the disease primes endothelial cells in vivo, which keep the acquired phenotype in culture.
Collapse
|
42
|
Kamimura D, Ohtani T, Sakata Y, Mano T, Takeda Y, Tamaki S, Omori Y, Tsukamoto Y, Furutani K, Komiyama Y, Yoshika M, Takahashi H, Matsuda T, Baba A, Umemura S, Miwa T, Komuro I, Yamamoto K. Ca2+ entry mode of Na+/Ca2+ exchanger as a new therapeutic target for heart failure with preserved ejection fraction. Eur Heart J 2011; 33:1408-16. [DOI: 10.1093/eurheartj/ehr106] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|