1
|
Pereira-Acácio A, Veloso-Santos JP, Silva-Rodrigues CO, Mello D, Alves-Bezerra DS, Costa-Sarmento G, Muzi-Filho H, Araújo-Silva CA, Lopes JA, Takiya CM, Cardozo SV, Vieyra A. Rostafuroxin, the inhibitor of endogenous ouabain, ameliorates chronic undernutrition-induced hypertension, stroke volume, cardiac output, left-ventricular fibrosis and alterations in Na +-transporting ATPases in rats. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2025; 11:100281. [PMID: 40182150 PMCID: PMC11967012 DOI: 10.1016/j.jmccpl.2024.100281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 04/05/2025]
Abstract
Our aim has been to investigate the effect of Rostafuroxin, an inhibitor of endogenous cardiotonic steroids (EO/CTS), on cardiac structure and function and cardiac Na+ transport in undernourished hypertensive Wistar rats, and to determine whether chronic undernutrition is a modifiable risk factor for cardiovascular-kidney-metabolic (CKM) syndrome. Echocardiographic studies evaluated stroke volume cardiac output, ejection fraction, mitral valve early diastolic blood flow/late diastolic blood flow (E/A) ratio, and right renal resistive index. The cardiomyocyte area and collagen infiltration of cardiac tissue were investigated, as also the activities of the cardiac ouabain-sensitive (Na++K+)ATPase ((Na++K+)ATPase Sens) and ouabain-resistant Na+-ATPase (Na+-ATPase Res). Undernourished hypertensive rats presented tachycardia, reduced stroke volume, decreased cardiac output, preserved fractional shortening and ejection fraction, unmodified mitral valve E/A ratio, and increased right renal resistive index. Cardiomyocyte size decreased and intense collagen infiltration had occurred. The (Na++K+)ATPase Sens activity decreased, whereas that of Na+-ATPase Res increased. Rostafuroxin selectively modified some of these echocardiographic and molecular parameters: it increased stroke volume and cardiac output and prevented histopathological alterations. The drug decreased and increased the activities of (Na++K+)ATPase Sens and Na+-ATPase Res, respectively, in normonourished rats, and the opposite trend was found in the undernourished group. It is concluded that chronic undernutrition in rats can provoke structural, functional, histological, and molecular cardiovascular alterations that, with the simultaneous changes in renal parameters described in this and in previous studies, configure an undescribed type of CKM syndrome. The data also demonstrate that the blockade of EO/CTS ameliorates stroke volume and cardiac output, thus preventing or delaying the worsening of the syndrome.
Collapse
Affiliation(s)
- Amaury Pereira-Acácio
- Graduate Program of Translational Biomedicine, Grande Rio University/UNIGRANRIO, Duque de Caxias, Brazil
| | - João P.M. Veloso-Santos
- Leopoldo de Meis Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Debora Mello
- National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danilo S. Alves-Bezerra
- Graduate Program of Translational Biomedicine, Grande Rio University/UNIGRANRIO, Duque de Caxias, Brazil
| | - Glória Costa-Sarmento
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Humberto Muzi-Filho
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlla A. Araújo-Silva
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jarlene A. Lopes
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Christina M. Takiya
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sergian V. Cardozo
- Graduate Program of Translational Biomedicine, Grande Rio University/UNIGRANRIO, Duque de Caxias, Brazil
- Grande Rio University/UNIGRANRIO, Duque de Caxias, Brazil
| | - Adalberto Vieyra
- Graduate Program of Translational Biomedicine, Grande Rio University/UNIGRANRIO, Duque de Caxias, Brazil
- National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
2
|
Socha MW, Chmielewski J, Pietrus M, Wartęga M. Endogenous Digitalis-like Factors as a Key Molecule in the Pathophysiology of Pregnancy-Induced Hypertension and a Potential Therapeutic Target in Preeclampsia. Int J Mol Sci 2023; 24:12743. [PMID: 37628922 PMCID: PMC10454430 DOI: 10.3390/ijms241612743] [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: 07/15/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Preeclampsia (PE), the most severe presentation of hypertensive disorders of pregnancy, is the major cause of morbidity and mortality linked to pregnancy, affecting both mother and fetus. Despite advances in prophylaxis and managing PE, delivery of the fetus remains the only causative treatment available. Focus on complex pathophysiology brought the potential for new treatment options, and more conservative options allowing reduction of feto-maternal complications and sequelae are being investigated. Endogenous digitalis-like factors, which have been linked to the pathogenesis of preeclampsia since the mid-1980s, have been shown to play a role in the pathogenesis of various cardiovascular diseases, including congestive heart failure and chronic renal disease. Elevated levels of EDLF have been described in pregnancy complicated by hypertensive disorders and are currently being investigated as a therapeutic target in the context of a possible breakthrough in managing preeclampsia. This review summarizes mechanisms implicating EDLFs in the pathogenesis of preeclampsia and evidence for their potential role in treating this doubly life-threatening disease.
Collapse
Affiliation(s)
- Maciej W. Socha
- Department of Perinatology, Gynecology and Gynecologic Oncology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland
- Department of Obstetrics and Gynecology, St. Adalbert’s Hospital in Gdańsk, Copernicus Healthcare Entity, Jana Pawła II 50, 80-462 Gdańsk, Poland
| | - Jakub Chmielewski
- Department of Obstetrics and Gynecology, St. Adalbert’s Hospital in Gdańsk, Copernicus Healthcare Entity, Jana Pawła II 50, 80-462 Gdańsk, Poland
| | - Miłosz Pietrus
- Department of Gynecology and Obstetrics, Jagiellonian University Medical College, 31-501 Kraków, Poland
| | - Mateusz Wartęga
- Department of Pathophysiology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie- Skłodowskiej 9, 85-094 Bydgoszcz, Poland
| |
Collapse
|
3
|
Carullo N, Fabiano G, D'Agostino M, Zicarelli MT, Musolino M, Presta P, Michael A, Andreucci M, Bolignano D, Coppolino G. New Insights on the Role of Marinobufagenin from Bench to Bedside in Cardiovascular and Kidney Diseases. Int J Mol Sci 2023; 24:11186. [PMID: 37446363 DOI: 10.3390/ijms241311186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Marinobufagenin (MBG) is a member of the bufadienolide family of compounds, which are natural cardiac glycosides found in a variety of animal species, including man, which have different physiological and biochemical functions but have a common action on the inhibition of the adenosine triphosphatase sodium-potassium pump (Na+/K+-ATPase). MBG acts as an endogenous cardiotonic steroid, and in the last decade, its role as a pathogenic factor in various human diseases has emerged. In this paper, we have collated major evidence regarding the biological characteristics and functions of MBG and its implications in human pathology. This review focused on MBG involvement in chronic kidney disease, including end-stage renal disease, cardiovascular diseases, sex and gender medicine, and its actions on the nervous and immune systems. The role of MBG in pathogenesis and the development of a wide range of pathological conditions indicate that this endogenous peptide could be used in the future as a diagnostic biomarker and/or therapeutic target, opening important avenues of scientific research.
Collapse
Affiliation(s)
- Nazareno Carullo
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Giuseppe Fabiano
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Mario D'Agostino
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | | | - Michela Musolino
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Pierangela Presta
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Ashour Michael
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Michele Andreucci
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Davide Bolignano
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Giuseppe Coppolino
- Renal Unit, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
4
|
Fedorova OV, Shilova VY, Zernetkina V, Juhasz O, Wei W, Lakatta EG, Bagrov AY. Silencing of PKG1 Gene Mimics Effect of Aging and Sensitizes Rat Vascular Smooth Muscle Cells to Cardiotonic Steroids: Impact on Fibrosis and Salt Sensitivity. J Am Heart Assoc 2023; 12:e028768. [PMID: 37301747 PMCID: PMC10356040 DOI: 10.1161/jaha.122.028768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/23/2023] [Indexed: 06/12/2023]
Abstract
Background Marinobufagenin, NKA (Na/K-ATPase) inhibitor, causes vasoconstriction and induces fibrosis via inhibition of Fli1 (Friend leukemia integration-1), a negative regulator of collagen synthesis. In vascular smooth muscle cells (VSMC), ANP (atrial natriuretic peptide), via a cGMP/PKG1 (protein kinase G1)-dependent mechanism, reduces NKA sensitivity to marinobufagenin. We hypothesized that VSMC from old rats, due to downregulation of ANP/cGMP/PKG-dependent signaling, would exhibit heightened sensitivity to the profibrotic effect of marinobufagenin. Methods and Results Cultured VSMC from the young (3-month-old) and old (24-month-old) male Sprague-Dawley rats and young VSMC with silenced PKG1 gene were treated with 1 nmol/L ANP, or with 1 nmol/L marinobufagenin, or with a combination of ANP and marinobufagenin. Collagen-1, Fli1, and PKG1 levels were assessed by Western blotting analyses. Vascular PKG1 and Fli1 levels in the old rats were reduced compared with their young counterparts. ANP prevented inhibition of vascular NKA by marinobufagenin in young VSMC but not in old VSMC. In VSMC from the young rats, marinobufagenin induced downregulation of Fli1 and an increase in collagen-1 level, whereas ANP blocked this effect. Silencing of the PKG1 gene in young VSMC resulted in a reduction in levels of PKG1 and Fli1; marinobufagenin additionally reduced Fli1 and increased collagen-1 level, and ANP failed to oppose these marinobufagenin effects, similar to VSMC from the old rats with the age-associated reduction in PKG1. Conclusions Age-associated reduction in vascular PKG1 and the resultant decline in cGMP signaling lead to the loss of the ability of ANP to oppose marinobufagenin-induced inhibition of NKA and fibrosis development. Silencing of the PKG1 gene mimicked these effects of aging.
Collapse
Affiliation(s)
- Olga V. Fedorova
- Laboratory of Cardiovascular ScienceNational Institute on Aging, NIHBaltimoreMDUSA
| | - Victoria Y. Shilova
- Laboratory of Cardiovascular ScienceNational Institute on Aging, NIHBaltimoreMDUSA
- Present address:
Engelhardt Institute of Molecular Biology, Russian Academy of SciencesMoscowRussia
| | - Valentina Zernetkina
- Laboratory of Cardiovascular ScienceNational Institute on Aging, NIHBaltimoreMDUSA
| | - Ondrej Juhasz
- Laboratory of Cardiovascular ScienceNational Institute on Aging, NIHBaltimoreMDUSA
| | - Wen Wei
- Laboratory of Cardiovascular ScienceNational Institute on Aging, NIHBaltimoreMDUSA
| | - Edward G. Lakatta
- Laboratory of Cardiovascular ScienceNational Institute on Aging, NIHBaltimoreMDUSA
| | - Alexei Y. Bagrov
- Laboratory of Cardiovascular ScienceNational Institute on Aging, NIHBaltimoreMDUSA
- Padakonn PharmaNarvaEstonia
| |
Collapse
|
5
|
Mukherji ST, Brambilla L, Stuart KB, Mayes I, Kutz LC, Chen Y, Barbosa LA, Elmadbouh I, McDermott JP, Haller ST, Romero MF, Soleimani M, Liu J, Shapiro JI, Blanco GV, Xie Z, Pierre SV. Na/K-ATPase signaling tonically inhibits sodium reabsorption in the renal proximal tubule. FASEB J 2023; 37:e22835. [PMID: 36856735 PMCID: PMC10028530 DOI: 10.1096/fj.202200785rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 03/02/2023]
Abstract
Through its classic ATP-dependent ion-pumping function, basolateral Na/K-ATPase (NKA) generates the Na+ gradient that drives apical Na+ reabsorption in the renal proximal tubule (RPT), primarily through the Na+ /H+ exchanger (NHE3). Accordingly, activation of NKA-mediated ion transport decreases natriuresis through activation of basolateral (NKA) and apical (NHE3) Na+ reabsorption. In contrast, activation of the more recently discovered NKA signaling function triggers cellular redistribution of RPT NKA and NHE3 and decreases Na+ reabsorption. We used gene targeting to test the respective contributions of NKA signaling and ion pumping to the overall regulation of RPT Na+ reabsorption. Knockdown of RPT NKA in cells and mice increased membrane NHE3 and Na+ /HCO3 - cotransporter (NBCe1A). Urine output and absolute Na+ excretion decreased by 65%, driven by increased RPT Na+ reabsorption (as indicated by decreased lithium clearance and unchanged glomerular filtration rate), and accompanied by elevated blood pressure. This hyper reabsorptive phenotype was rescued upon crossing with RPT NHE3-/- mice, confirming the importance of NKA/NHE3 coupling. Hence, NKA signaling exerts a tonic inhibition on Na+ reabsorption by regulating key apical and basolateral Na+ transporters. This action, lifted upon NKA genetic suppression, tonically counteracts NKA's ATP-driven function of basolateral Na+ reabsorption. Strikingly, NKA signaling is not only physiologically relevant but it also appears to be functionally dominant over NKA ion pumping in the control of RPT reabsorption.
Collapse
Affiliation(s)
- Shreya T. Mukherji
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Luca Brambilla
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Kailey B. Stuart
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Isabella Mayes
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Laura C. Kutz
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Yiliang Chen
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Blood Research Institute, Versiti, WI
| | - Leandro A Barbosa
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
- Universidade Federal de São João del-Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Ibrahim Elmadbouh
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Jeff P. McDermott
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS
| | - Steven T. Haller
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Michael F. Romero
- Physiology & Biomedical Engineering and Nephrology & Hypertension, Mayo Clinic College of Medicine & Science, Rochester, MN
| | - Manoocher Soleimani
- Department of Medicine, The University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Jiang Liu
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | - Joseph I. Shapiro
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | - Gustavo V. Blanco
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Sandrine V. Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| |
Collapse
|
6
|
Lamichhane S, Mohammed CJ, Haller ST, Kennedy DJ, Isailovic D. Quantification of Cardiotonic Steroids Potentially Regulated by Paraoxonase 3 in a Rat Model of Chronic Kidney Disease Using UHPLC-Orbitrap-MS. Int J Mol Sci 2022; 23:ijms232113565. [PMID: 36362352 PMCID: PMC9654389 DOI: 10.3390/ijms232113565] [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: 09/21/2022] [Revised: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Endogenous cardiotonic steroids (CTSs), such as telocinobufagin (TCB) and marinobufagin (MBG) contain a lactone moiety critical to their binding and signaling through the Na+/K+-ATPase. Their concentrations elevate in response to sodium intake and under volume-expanded conditions. Paraoxonase 3 (PON3) is an enzyme that can hydrolyze lactone substrates. Here, we examine the role of PON3 in regulating CTS levels in a rat model of chronic kidney diseases (CKD). TCB and MBG were extracted from rat urine samples, and the analyses were carried out using ultra-high pressure liquid chromatography−Orbitrap-mass spectrometry (UHPLC-Orbitrap-MS). Ten-week-old Dahl salt-sensitive wild type (SS-WT) and Dahl salt-sensitive PON3 knockout (SS-PON3 KO) rats were maintained on a high-salt diet (8% NaCl) for 8 weeks to initiate salt-sensitive hypertensive renal disease characteristic of this model. CTS extraction recovery from urine >80% was achieved. For animals maintained on a normal chow diet, the baseline amount of TCB excreted in 24 h urine of SS-PON3 KO rats (6.08 ± 1.47 ng/24 h; or 15.09 ± 3.25 pmol) was significantly higher than for SS-WT rats (1.48 ± 0.69 ng/24 h; or 3.67 ± 1.54 pmol, p < 0.05). Similarly, for the same animals, the amount of excreted MBG was higher in the urine of SS-PON3 KO rats (4.74 ± 1.30 ng/24 h versus 1.03 ± 0.25 ng/24 h in SS-WT; or 11.83 ± 2.91 pmol versus 2.57 ± 0.56 pmol in SS-WT, p < 0.05). For animals on a high-salt diet, the SS-PON3 KO rats had significantly increased levels of TCB (714.52 ± 79.46 ng/24 h; or 1774.85 ± 175.55 pmol) compared to SS-WT control (343.84 ± 157.54 ng/24 h; or 854.09 ± 350.02 pmol, p < 0.05), and comparatively higher levels of MBG were measured for SS-PON3 KO (225.55 ± 82.61 ng/24 h; or 563.19 ± 184.5 pmol) versus SS-WT (157.56 ± 85.53 ng/24 h; or 393.43 ± 191.01 pmol, p > 0.05) rats. These findings suggest that the presence and absence of PON3 dramatically affect the level of endogenous CTSs, indicating its potential role in CTS regulation.
Collapse
Affiliation(s)
- Sabitri Lamichhane
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - Chrysan J. Mohammed
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Steven T. Haller
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - David J. Kennedy
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
- Correspondence: (D.J.K.); (D.I.); Tel.: +1+419-383-6822 (D.J.K.); +1+419-530-5523 (D.I.)
| | - Dragan Isailovic
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
- Correspondence: (D.J.K.); (D.I.); Tel.: +1+419-383-6822 (D.J.K.); +1+419-530-5523 (D.I.)
| |
Collapse
|
7
|
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
|
8
|
Abdelmissih S, Sayed WM, Rashed LA, Kamel MM, Eshra MA, Attallah MI, El-Naggar RAR. The extent of involvement of ouabain, hippocampal expression of Na+/K+-ATPase, and corticosterone/melatonin receptors ratio in modifying stress-induced behavior differs according to the stressor in context. Braz J Med Biol Res 2022; 55:e11938. [PMID: 35857994 PMCID: PMC9296128 DOI: 10.1590/1414-431x2022e11938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 04/18/2022] [Indexed: 11/21/2022] Open
Abstract
The aim of this study was to assess the effect of two types of stressors,
regarding the extent of involvement of ouabain (OUA), hippocampal
sodium/potassium ATPase (NKA) expression, and the hippocampal
corticosterone receptors (CR)/melatonin receptors
(MR) expression ratio, on the behavioral and cardiovascular
responses and on the hippocampal cornu ammonis zone 3 (CA3) and dentate gyrus
(DG). Thirty adult male Wistar albino rats aged 7-8 months were exposed to
either chronic immobilization or a disturbed dark/light cycle and treated with
either ouabain or vehicle. In the immobilized group, in the absence of
hippocampal corticosterone (CORT) changes, rats were non-responsive to stress,
despite experiencing increased pulse rate, downregulated hippocampal
sodium/potassium pump, and enhanced hippocampal CR/MR
expression ratio. Prolonged darkness precipitated a reduced upright attack
posture, with elevated CORT against hippocampal MR
downregulation. Both immobilization and, to a lesser extent, prolonged darkness
stress resulted in histopathological and ultrastructural neurodegenerative
changes in the hippocampus. OUA administration did not change the behavioral
resilience in restrained rats, despite persistence of the underlying biochemical
derangements, added to decreased CORT. On the contrary, with exposure to short
photoperiods, OUA reverted the behavior towards a combative reduction of
inactivity, with unvaried CR/MR and CORT, while ameliorating
hippocampal neuro-regeneration, with co-existing NKA and
MR repressions. Therefore, the extent of OUA, hippocampal
NKA expression, and CR/MR expression, and
subsequent behavioral and cardiac responses and hippocampal histopathology,
differ according to the type of stressor, whether immobilization or prolonged
darkness.
Collapse
Affiliation(s)
- S Abdelmissih
- Department of Medical Pharmacology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt
| | - W M Sayed
- Department of Anatomy and Embryology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt
| | - L A Rashed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt
| | - M M Kamel
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt.,Department of Basic Medical Science, Faculty of Medicine, King Salman International University, South Sinai, Egypt
| | - M A Eshra
- Department of Physiology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt
| | - M I Attallah
- Department of Medical Pharmacology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt
| | - R A-R El-Naggar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr University for Science and Technology, Giza, Egypt
| |
Collapse
|
9
|
Grigorova YN, Juhasz O, Long JM, Zernetkina VI, Hall ML, Wei W, Morrell CH, Petrashevskaya N, Morrow A, LaNasa KH, Bagrov AY, Rapp PR, Lakatta EG, Fedorova OV. Effect of Cardiotonic Steroid Marinobufagenin on Vascular Remodeling and Cognitive Impairment in Young Dahl-S Rats. Int J Mol Sci 2022; 23:4563. [PMID: 35562955 PMCID: PMC9101263 DOI: 10.3390/ijms23094563] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/17/2022] [Accepted: 04/17/2022] [Indexed: 02/04/2023] Open
Abstract
The hypertensive response in Dahl salt-sensitive (DSS) rats on a high-salt (HS) diet is accompanied by central arterial stiffening (CAS), a risk factor for dementia, and heightened levels of a prohypertensive and profibrotic factor, the endogenous Na/K-ATPase inhibitor marinobufagenin (MBG). We studied the effect of the in vivo administration of MBG or HS diet on blood pressure (BP), CAS, and behavioral function in young DSS rats and normotensive Sprague-Dawley rats (SD), the genetic background for DSS rats. Eight-week-old male SD and DSS rats were given an HS diet (8% NaCl, n = 18/group) or a low-salt diet (LS; 0.1% NaCl, n = 14-18/group) for 8 weeks or MBG (50 µg/kg/day, n = 15-18/group) administered via osmotic minipumps for 4 weeks in the presence of the LS diet. The MBG-treated groups received the LS diet. The systolic BP (SBP); the aortic pulse wave velocity (aPWV), a marker of CAS; MBG levels; spatial memory, measured by a water maze task; and tissue collection for the histochemical analysis were assessed at the end of the experiment. DSS-LS rats had higher SBP, higher aPWV, and poorer spatial memory than SD-LS rats. The administration of stressors HS and MBG increased aPWV, SBP, and aortic wall collagen abundance in both strains vs. their LS controls. In SD rats, HS or MBG administration did not affect heart parameters, as assessed by ECHO vs. the SD-LS control. In DSS rats, impaired whole-heart structure and function were observed after HS diet administration in DSS-HS vs. DSS-LS rats. MBG treatment did not affect the ECHO parameters in DSS-MBG vs. DSS-LS rats. The HS diet led to an increase in endogenous plasma and urine MBG levels in both SD and DSS groups. Thus, the prohypertensive and profibrotic effect of HS diet might be partially attributed to an increase in MBG. The prohypertensive and profibrotic functions of MBG were pronounced in both DSS and SD rats, although quantitative PCR revealed that different profiles of profibrotic genes in DSS and SD rats was activated after MBG or HS administration. Spatial memory was not affected by HS diet or MBG treatment in either SD or DSS rats. Impaired cognitive function was associated with higher BP, CAS, and cardiovascular remodeling in young DSS-LS rats, as compared to young SD-LS rats. MBG and HS had similar effects on the cardiovascular system and its function in DSS and SD rats, although the rate of change in SD rats was lower than in DSS rats. The absence of a cumulative effect of increased aPWV and BP on spatial memory can be explained by the cerebrovascular and brain plasticity in young rats, which help the animals to tolerate CAS elevated by HS and MBG and to counterbalance the profibrotic effect of heightened MBG.
Collapse
Affiliation(s)
- Yulia N. Grigorova
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA; (Y.N.G.); (O.J.); (V.I.Z.); (M.L.H.); (W.W.); (C.H.M.); (N.P.); (A.Y.B.); (E.G.L.)
| | - Ondrej Juhasz
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA; (Y.N.G.); (O.J.); (V.I.Z.); (M.L.H.); (W.W.); (C.H.M.); (N.P.); (A.Y.B.); (E.G.L.)
| | - Jeffrey M. Long
- Laboratory of Behavioral Neuroscience, Neurocognitive Aging Section, National Institute on Aging, NIH, Baltimore, MD 21224, USA; (J.M.L.); (A.M.); (K.H.L.); (P.R.R.)
| | - Valentina I. Zernetkina
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA; (Y.N.G.); (O.J.); (V.I.Z.); (M.L.H.); (W.W.); (C.H.M.); (N.P.); (A.Y.B.); (E.G.L.)
| | - Mikayla L. Hall
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA; (Y.N.G.); (O.J.); (V.I.Z.); (M.L.H.); (W.W.); (C.H.M.); (N.P.); (A.Y.B.); (E.G.L.)
| | - Wen Wei
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA; (Y.N.G.); (O.J.); (V.I.Z.); (M.L.H.); (W.W.); (C.H.M.); (N.P.); (A.Y.B.); (E.G.L.)
| | - Christopher H. Morrell
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA; (Y.N.G.); (O.J.); (V.I.Z.); (M.L.H.); (W.W.); (C.H.M.); (N.P.); (A.Y.B.); (E.G.L.)
| | - Natalia Petrashevskaya
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA; (Y.N.G.); (O.J.); (V.I.Z.); (M.L.H.); (W.W.); (C.H.M.); (N.P.); (A.Y.B.); (E.G.L.)
| | - Audrey Morrow
- Laboratory of Behavioral Neuroscience, Neurocognitive Aging Section, National Institute on Aging, NIH, Baltimore, MD 21224, USA; (J.M.L.); (A.M.); (K.H.L.); (P.R.R.)
| | - Katherine H. LaNasa
- Laboratory of Behavioral Neuroscience, Neurocognitive Aging Section, National Institute on Aging, NIH, Baltimore, MD 21224, USA; (J.M.L.); (A.M.); (K.H.L.); (P.R.R.)
| | - Alexei Y. Bagrov
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA; (Y.N.G.); (O.J.); (V.I.Z.); (M.L.H.); (W.W.); (C.H.M.); (N.P.); (A.Y.B.); (E.G.L.)
| | - Peter R. Rapp
- Laboratory of Behavioral Neuroscience, Neurocognitive Aging Section, National Institute on Aging, NIH, Baltimore, MD 21224, USA; (J.M.L.); (A.M.); (K.H.L.); (P.R.R.)
| | - Edward G. Lakatta
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA; (Y.N.G.); (O.J.); (V.I.Z.); (M.L.H.); (W.W.); (C.H.M.); (N.P.); (A.Y.B.); (E.G.L.)
| | - Olga V. Fedorova
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA; (Y.N.G.); (O.J.); (V.I.Z.); (M.L.H.); (W.W.); (C.H.M.); (N.P.); (A.Y.B.); (E.G.L.)
| |
Collapse
|
10
|
Liu J, Tian J, Sodhi K, Shapiro JI. The Na/K-ATPase Signaling and SGLT2 Inhibitor-Mediated Cardiorenal Protection: A Crossed Road? J Membr Biol 2021; 254:513-529. [PMID: 34297135 PMCID: PMC8595165 DOI: 10.1007/s00232-021-00192-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/06/2021] [Indexed: 12/17/2022]
Abstract
In different large-scale clinic outcome trials, sodium (Na+)/glucose co-transporter 2 (SGLT2) inhibitors showed profound cardiac- and renal-protective effects, making them revolutionary treatments for heart failure and kidney disease. Different theories are proposed according to the emerging protective effects other than the original purpose of glucose-lowering in diabetic patients. As the ATP-dependent primary ion transporter providing the Na+ gradient to drive other Na+-dependent transporters, the possible role of the sodium–potassium adenosine triphosphatase (Na/K-ATPase) as the primary ion transporter and its signaling function is not explored.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Biomedical Sciences, JCE School of Medicine, Marshall University, Huntington, WV, USA.
| | - Jiang Tian
- Department of Biomedical Sciences, JCE School of Medicine, Marshall University, Huntington, WV, USA
| | - Komal Sodhi
- Department of Surgery, JCE School of Medicine, Marshall University, Huntington, WV, USA
| | - Joseph I Shapiro
- Departments of Medicine, JCE School of Medicine, Marshall University, Huntington, WV, USA
| |
Collapse
|
11
|
McCarty MF. Nutraceutical, Dietary, and Lifestyle Options for Prevention and Treatment of Ventricular Hypertrophy and Heart Failure. Int J Mol Sci 2021; 22:ijms22073321. [PMID: 33805039 PMCID: PMC8037104 DOI: 10.3390/ijms22073321] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Although well documented drug therapies are available for the management of ventricular hypertrophy (VH) and heart failure (HF), most patients nonetheless experience a downhill course, and further therapeutic measures are needed. Nutraceutical, dietary, and lifestyle measures may have particular merit in this regard, as they are currently available, relatively safe and inexpensive, and can lend themselves to primary prevention as well. A consideration of the pathogenic mechanisms underlying the VH/HF syndrome suggests that measures which control oxidative and endoplasmic reticulum (ER) stress, that support effective nitric oxide and hydrogen sulfide bioactivity, that prevent a reduction in cardiomyocyte pH, and that boost the production of protective hormones, such as fibroblast growth factor 21 (FGF21), while suppressing fibroblast growth factor 23 (FGF23) and marinobufagenin, may have utility for preventing and controlling this syndrome. Agents considered in this essay include phycocyanobilin, N-acetylcysteine, lipoic acid, ferulic acid, zinc, selenium, ubiquinol, astaxanthin, melatonin, tauroursodeoxycholic acid, berberine, citrulline, high-dose folate, cocoa flavanols, hawthorn extract, dietary nitrate, high-dose biotin, soy isoflavones, taurine, carnitine, magnesium orotate, EPA-rich fish oil, glycine, and copper. The potential advantages of whole-food plant-based diets, moderation in salt intake, avoidance of phosphate additives, and regular exercise training and sauna sessions are also discussed. There should be considerable scope for the development of functional foods and supplements which make it more convenient and affordable for patients to consume complementary combinations of the agents discussed here. Research Strategy: Key word searching of PubMed was employed to locate the research papers whose findings are cited in this essay.
Collapse
Affiliation(s)
- Mark F McCarty
- Catalytic Longevity Foundation, 811 B Nahant Ct., San Diego, CA 92109, USA
| |
Collapse
|
12
|
Gohar EY, Almutlaq RN, Daugherty EM, Butt MK, Jin C, Pollock JS, Pollock DM, De Miguel C. Activation of G protein-coupled estrogen receptor 1 ameliorates proximal tubular injury and proteinuria in Dahl salt-sensitive female rats. Am J Physiol Regul Integr Comp Physiol 2021; 320:R297-R306. [PMID: 33407017 PMCID: PMC7988769 DOI: 10.1152/ajpregu.00267.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/21/2020] [Accepted: 12/21/2020] [Indexed: 01/02/2023]
Abstract
Recent evidence indicates a crucial role for G protein-coupled estrogen receptor 1 (GPER1) in the maintenance of cardiovascular and kidney health in females. The current study tested whether GPER1 activation ameliorates hypertension and kidney damage in female Dahl salt-sensitive (SS) rats fed a high-salt (HS) diet. Adult female rats were implanted with telemetry transmitters for monitoring blood pressure and osmotic minipumps releasing G1 (selective GPER1 agonist, 400 μg/kg/day ip) or vehicle. Two weeks after pump implantation, rats were shifted from a normal-salt (NS) diet (0.4% NaCl) to a matched HS diet (4.0% NaCl) for 2 wk. Twenty-four hour urine samples were collected during both diet periods and urinary markers of kidney injury were assessed. Histological assessment of kidney injury was conducted after the 2-wk HS diet period. Compared with values during the NS diet, 24-h mean arterial pressure markedly increased in response to HS, reaching similar values in vehicle-treated and G1-treated rats. HS also significantly increased urinary excretion of protein, albumin, nephrin (podocyte damage marker), and KIM-1 (proximal tubule injury marker) in vehicle-treated rats. Importantly, G1 treatment prevented the HS-induced proteinuria, albuminuria, and increase in KIM-1 excretion but not nephrinuria. Histological analysis revealed that HS-induced glomerular damage did not differ between groups. However, G1 treatment preserved proximal tubule brush-border integrity in HS-fed rats. Collectively, our data suggest that GPER1 activation protects against HS-induced proteinuria and albuminuria in female Dahl SS rats by preserving proximal tubule brush-border integrity in a blood pressure-independent manner.
Collapse
Affiliation(s)
- Eman Y Gohar
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Rawan N Almutlaq
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Elizabeth M Daugherty
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Maryam K Butt
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Chunhua Jin
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jennifer S Pollock
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - David M Pollock
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Carmen De Miguel
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
13
|
Hassan MJM, Bakar NS, Aziz MA, Basah NK, Singh HJ. Leptin-induced increase in blood pressure and markers of endothelial activation during pregnancy in Sprague Dawley rats is prevented by resibufogenin, a marinobufagenin antagonist. Reprod Biol 2020; 20:184-190. [PMID: 32253169 DOI: 10.1016/j.repbio.2020.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/06/2020] [Accepted: 03/14/2020] [Indexed: 11/26/2022]
Abstract
Levels of leptin and marinobufagenin (MBG), a cardiotonic steroid, are elevated in the serum of women with pre-eclampsia. Besides this, leptin administration to pregnant rats increases systolic blood pressure (SBP), urinary protein excretion and serum markers of endothelial activation. The link between leptin and MBG is unknown and it is also unclear if leptin-induced increases in blood pressure and proteinuria in the pregnant rat could be prevented by an MBG antagonist. To ascertain this link, this study investigated the effect of resibufogenin (RBG), a marinobufagenin antagonist, on leptin-induced increases in blood pressure and proteinuria during pregnancy in rats. Four groups of Sprague-Dawley rats, aged 12 weeks, were given either normal saline (CONTROL) or 120 μg/kg/day of leptin (LEP), or 120 μg/kg/day of leptin+30 μg/kg/day of resibufogenin (L + RBG) or 30 μg/kg/day of resibufogenin (RBG) from Day 1-20 of pregnancy. Systolic blood pressure and urinary protein excretion (UPE) were measured during the study period. Animals were euthanized on day 21 of pregnancy and vascular cell adhesion molecule 1, (VCAM-1), soluble intracellular cell adhesion molecule 1 (sICAM-1), E-selectin and endothelin-1 (ET-1) were estimated in the serum. SBP, UPE, VCAM-1, sICAM-1 and ET-1 were significantly higher only in the LEP group when compared with those in CONT and in L + RBG and RBG groups. The prevention by RBG of leptin-induced increases in SBP, proteinuria, and endothelial activation during pregnancy seem to suggest a potential role for MBG in leptin-induced adverse effects on blood pressure, urinary protein excretion and endothelial activity during pregnancy in the rat.
Collapse
Affiliation(s)
| | - Nor Salmah Bakar
- Faculty of Medicine, Universiti Teknologi MARA Sungai Buloh Campus, Selangor, Malaysia
| | - Mardiana Abdul Aziz
- Faculty of Medicine, Universiti Teknologi MARA Sungai Buloh Campus, Selangor, Malaysia
| | - Norizan Kamal Basah
- Faculty of Medicine, Universiti Teknologi MARA Sungai Buloh Campus, Selangor, Malaysia
| | - Harbindar Jeet Singh
- Faculty of Medicine, Universiti Teknologi MARA Sungai Buloh Campus, Selangor, Malaysia; I-PPerFORM, Universiti Teknologi MARA, Malaysia.
| |
Collapse
|
14
|
Orlov SN, Tverskoi AM, Sidorenko SV, Smolyaninova LV, Lopina OD, Dulin NO, Klimanova EA. Na,K-ATPase as a target for endogenous cardiotonic steroids: What's the evidence? Genes Dis 2020; 8:259-271. [PMID: 33997173 PMCID: PMC8093582 DOI: 10.1016/j.gendis.2020.01.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/24/2019] [Accepted: 01/09/2020] [Indexed: 12/17/2022] Open
Abstract
With an exception of few reports, the plasma concentration of ouabain and marinobufagenin, mostly studied cardiotonic steroids (CTS) assessed by immunoassay techniques, is less than 1 nM. During the last 3 decades, the implication of these endogenous CTS in the pathogenesis of hypertension and other volume-expanded disorders is widely disputed. The threshold for inhibition by CTS of human and rodent α1-Na,K-ATPase is ∼1 and 1000 nM, respectively, that rules out the functioning of endogenous CTS (ECTS) as natriuretic hormones and regulators of cell adhesion, cell-to-cell communication, gene transcription and translation, which are mediated by dissipation of the transmembrane gradients of monovalent cations. In several types of cells ouabain and marinobufagenin at concentrations corresponding to its plasma level activate Na,K-ATPase, decrease the [Na+]i/[K+]i-ratio and increase cell proliferation. Possible physiological significance and mechanism of non-canonical Na+i/K+i-dependent and Na+i/K+i-independent cell responses to CTS are discussed.
Collapse
Affiliation(s)
- Sergei N Orlov
- MV Lomonosov Moscow State University, Moscow, 119234, Russia.,National Research Tomsk State University, Tomsk, 634050, Russia.,Siberian State Medical University, Tomsk, 634050, Russia
| | | | - Svetlana V Sidorenko
- MV Lomonosov Moscow State University, Moscow, 119234, Russia.,National Research Tomsk State University, Tomsk, 634050, Russia
| | - Larisa V Smolyaninova
- MV Lomonosov Moscow State University, Moscow, 119234, Russia.,National Research Tomsk State University, Tomsk, 634050, Russia
| | - Olga D Lopina
- MV Lomonosov Moscow State University, Moscow, 119234, Russia
| | | | - Elizaveta A Klimanova
- MV Lomonosov Moscow State University, Moscow, 119234, Russia.,National Research Tomsk State University, Tomsk, 634050, Russia
| |
Collapse
|
15
|
Pavlovic D. Endogenous cardiotonic steroids and cardiovascular disease, where to next? Cell Calcium 2019; 86:102156. [PMID: 31896530 PMCID: PMC7031694 DOI: 10.1016/j.ceca.2019.102156] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 12/24/2019] [Accepted: 12/24/2019] [Indexed: 11/18/2022]
Abstract
Ever since British Physician William Withering first described the use of foxglove extract for treatment of patients with congestive heart failure in 1785, cardiotonic steroids have been used clinically to treat heart failure and more recently atrial fibrillation. Due to their ability to bind and inhibit the ubiquitous transport enzyme sodium potassium pump, thus regulating intracellular Na+ concentration in every living cell, they are also an essential tool for research into the sodium potassium pump structure and function. Exogenous CTS have been clearly demonstrated to affect cardiovascular system through modulation of vagal tone, cardiac contraction (via ionic changes) and altered natriuresis. Reports of a number of endogenous CTS, since the 1980s, have intensified research into their physiologic and pathophysiologic roles and opened up novel therapeutic targets. Substantive evidence pointing to the role of endogenous ouabain and marinobufagenin, the two most prominent CTS, in development of cardiovascular disease has accumulated. Nevertheless, their presence, structure, biosynthesis pathways and even mechanism of action remain unclear or controversial. In this review the current state-of-the-art, the controversies and the remaining questions surrounding the role of endogenous cardiotonic steroids in health and disease are discussed.
Collapse
Affiliation(s)
- Davor Pavlovic
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
16
|
Strauss-Kruger M, Smith W, Wei W, Bagrov AY, Fedorova OV, Schutte AE. Microvascular function in non-dippers: Potential involvement of the salt sensitivity biomarker, marinobufagenin-The African-PREDICT study. J Clin Hypertens (Greenwich) 2019; 22:86-94. [PMID: 31873989 DOI: 10.1111/jch.13767] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/20/2019] [Accepted: 12/02/2019] [Indexed: 12/01/2022]
Abstract
Suppressed nighttime blood pressure dipping is associated with salt sensitivity and may increase the hemodynamic load on the microvasculature. The mechanism remains unknown whereby salt sensitivity may increase the cardiovascular risk of non-dippers. Marinobufagenin, a novel steroidal biomarker, is associated with salt sensitivity and other cardiovascular risk factors independent of blood pressure. The authors investigated whether microvascular function in non-dippers is associated with marinobufagenin. The authors included 220 dippers and 154 non-dippers (aged 20-30 years) from the African-PREDICT study, with complete 24-hour urinary marinobufagenin and sodium data. The authors determined dipping status using 24-hour blood pressure monitoring and defined nighttime non-dipping <10%. The authors measured microvascular reactivity as retinal artery dilation in response to light flicker provocation. Young healthy non-dippers and dippers presented with similar peak retinal artery dilation, urinary sodium, and MBG excretion (P > .05). However, only in non-dippers did peak retinal artery dilation relate negatively to marinobufagenin excretion after single (r = -0.20; P = .012), partial (r = -0.23; P = .004), and multivariate-adjusted regression analyses (Adj. R2 = 0.34; β = -0.26; P < .001). The authors also noted a relationship between peak artery dilation and estimated salt intake (Adj. R2 = 0.30; β = -0.14; P = .051), but it was lost upon inclusion of marinobufagenin (Adj. R2 = 0.33; β = -0.015; P = .86). No relationship between microvascular reactivity and marinobufagenin was evident in dippers (P = .77). Marinobufagenin, representing salt sensitivity, may be involved in early microvascular functional changes in young non-dippers and thus contributes to the development of hypertension and cardiovascular disease later in life.
Collapse
Affiliation(s)
- Michél Strauss-Kruger
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
| | - Wayne Smith
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa.,Hypertension and Cardiovascular Disease, MRC Research Unit, North-West University, Potchefstroom, South Africa
| | - Wen Wei
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Alexei Y Bagrov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, St. Petersburg, Russia
| | - Olga V Fedorova
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Aletta E Schutte
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa.,Hypertension and Cardiovascular Disease, MRC Research Unit, North-West University, Potchefstroom, South Africa
| |
Collapse
|
17
|
Potent nonopioid antinociceptive activity of telocinobufagin in models of acute pain in mice. Pain Rep 2019; 4:e791. [PMID: 31984296 PMCID: PMC6903372 DOI: 10.1097/pr9.0000000000000791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/04/2019] [Accepted: 09/03/2019] [Indexed: 11/27/2022] Open
Abstract
Introduction: In recent decades, several researches have been conducted in search of new analgesics that do not present the side effects of opioids. In this context, animal venoms contain natural painkillers that have been used for the development of new analgesics. Objective: The aims of this study were to evaluate the antinociceptive effects of telocinobufagin (TCB), a bufadienolide isolated from Rhinella jimi venom, in murine acute pain models, and to verify the participation of the opioid system in these effects. Methods: TCB was purified from R. jimi venom by high-performance liquid chromatography, and its structure was confirmed by spectrometric techniques. TCB was administered intraperitoneally (i.p.) (0.062, 0.125, 0.25, 0.5, and 1 mg·kg−1) and orally (p.o.) (0.625, 1.125, 2.5, 5, and 10 mg·kg−1) in mice, which were then subjected to pain tests: acetic acid–induced writhing, formalin, tail-flick, and hot-plate. Involvement of the opioid system in TCB action was evaluated by naloxone i.p. injected (2.5 mg·kg−1) 20 minutes before TCB administration. In addition, the TCB action on the μ, δ, and κ opioid receptors was performed by radioligand binding assays. Results: In all the tests used, TCB showed dose-dependent antinociceptive activity with more than 90% inhibition of the nociceptive responses at the doses of 1 mg·kg−1 (i.p.) and 10 mg·kg−1 (p.o.). Naloxone did not alter the effect of TCB. In addition, TCB did not act on the μ, δ, and κ opioid receptors. Conclusion: The results suggest that TCB may represent a novel potential nonopioid therapeutic analgesic for treatment of acute pains.
Collapse
|
18
|
Zhang Y, Wei W, Shilova V, Petrashevskaya NN, Zernetkina VI, Grigorova YN, Marshall CA, Fenner RC, Lehrmann E, Wood WH, Becker KG, Lakatta EG, Bagrov AY, Fedorova OV. Monoclonal Antibody to Marinobufagenin Downregulates TGFβ Profibrotic Signaling in Left Ventricle and Kidney and Reduces Tissue Remodeling in Salt-Sensitive Hypertension. J Am Heart Assoc 2019; 8:e012138. [PMID: 31576777 PMCID: PMC6818028 DOI: 10.1161/jaha.119.012138] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background Elevated levels of an endogenous Na/K-ATPase inhibitor marinobufagenin accompany salt-sensitive hypertension and are implicated in cardiac fibrosis. Immunoneutralization of marinobufagenin reduces blood pressure in Dahl salt-sensitive (Dahl-S) rats. The effect of the anti-marinobufagenin monoclonal antibody on blood pressure, left ventricular (LV) and renal remodeling, and gene expression were investigated in hypertensive Dahl-S rats. Methods and Results Dahl-S rats were fed high NaCl (8%, HS; n=14) or low NaCl (0.1%, LS; n=14) diets for 8 weeks. Animals were administered control antibody (LS control antibody, LSC; HS control antibody, HSC; n=7 per group) or anti-marinobufagenin antibody once on week 7 of diet intervention (n=7 per group). Levels of marinobufagenin, LV, and kidney mRNAs and proteins implicated in profibrotic signaling were assessed. Systolic blood pressure was elevated (211±8 versus 133±3 mm Hg, P<0.01), marinobufagenin increased 2-fold in plasma (P<0.05) and 5-fold in urine (P<0.01), LV and kidney weights increased, and levels of LV collagen-1 rose 3.5-fold in HSC versus LSC. Anti-marinobufagenin antibody treatment decreased systolic blood pressure by 24 mm Hg (P<0.01) and reduced organ weights and level of LV collagen-1 (P<0.01) in hypertensive Dahl salt-sensitive rats with anti-marinobufagenin antibody versus HSC. The expression of genes related to transforming growth factor-β-dependent signaling was upregulated in the left ventricles and kidneys in HSC versus LSC groups and became downregulated following administration of anti-marinobufagenin antibody to hypertensive Dahl-S rats. Marinobufagenin also activated transforming growth factor-β signaling in cultured ventricular myocytes from Dahl-S rats. Conclusions Immunoneutralization of heightened marinobufagenin levels in hypertensive Dahl-S rats resulted in a downregulation of genes implicated in transforming growth factor-β pathway, which indicates that marinobufagenin is an activator of profibrotic transforming growth factor-β-dependent signaling in salt-sensitive hypertension.
Collapse
Affiliation(s)
- Yongqing Zhang
- Laboratory of Genetics and Genomics National Institute on Aging NIH Baltimore MD
| | - Wen Wei
- Laboratory of Cardiovascular Science National Institute on Aging NIH Baltimore MD
| | - Victoria Shilova
- Laboratory of Cardiovascular Science National Institute on Aging NIH Baltimore MD
| | | | | | - Yulia N Grigorova
- Laboratory of Cardiovascular Science National Institute on Aging NIH Baltimore MD
| | - Courtney A Marshall
- Laboratory of Cardiovascular Science National Institute on Aging NIH Baltimore MD
| | - Rachel C Fenner
- Laboratory of Cardiovascular Science National Institute on Aging NIH Baltimore MD
| | - Elin Lehrmann
- Laboratory of Genetics and Genomics National Institute on Aging NIH Baltimore MD
| | - William H Wood
- Laboratory of Genetics and Genomics National Institute on Aging NIH Baltimore MD
| | - Kevin G Becker
- Laboratory of Genetics and Genomics National Institute on Aging NIH Baltimore MD
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science National Institute on Aging NIH Baltimore MD
| | - Alexei Y Bagrov
- Laboratory of Cardiovascular Science National Institute on Aging NIH Baltimore MD
| | - Olga V Fedorova
- Laboratory of Cardiovascular Science National Institute on Aging NIH Baltimore MD
| |
Collapse
|
19
|
Marinobufagenin Inhibits Neutrophil Migration and Proinflammatory Cytokines. J Immunol Res 2019; 2019:1094520. [PMID: 31236418 PMCID: PMC6545758 DOI: 10.1155/2019/1094520] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/28/2019] [Indexed: 12/12/2022] Open
Abstract
Cardiotonic steroids, such as ouabain and digoxin, are known to bind to Na+/K+-ATPase and to promote several biological activities, including anti-inflammatory activity. However, there are still no reports in the literature about inflammation and marinobufagenin, a cardiotonic steroid from the bufadienolide family endogenously found in mammals. Therefore, the aim of this work was to analyze, in vivo and in vitro, the role of marinobufagenin in acute inflammation. Swiss mice were treated with 0.56 mg/kg of marinobufagenin intraperitoneally (i.p.) and zymosan (2 mg/mL, i.p.) was used to induce peritoneal inflammation. Peritoneal fluid was collected and used for counting cells by optical microscopy and proinflammatory cytokine quantification (IL-1β, IL-6, and TNF-α) by immunoenzymatic assay (ELISA). Zymosan stimulation, as expected, induced increased cell migration and proinflammatory cytokine levels in the peritoneum. Marinobufagenin treatment reduced polymorphonuclear cell migration and IL-1β and IL-6 levels in the peritoneal cavity, without interfering in TNF-α levels. In addition, the effect of marinobufagenin was evaluated using peritoneal macrophages stimulated by zymosan (0.2 mg/mL) in vitro. Marinobufagenin treatment at different concentrations (10, 100, 1000, and 10000 nM) showed no cytotoxic effect on peritoneal macrophages. Interestingly, the lowest concentration, which did not inhibit Na+/K+-ATPase activity, attenuated proinflammatory cytokines IL-1β, IL-6, and TNF-α levels. To investigate the putative mechanism of action of marinobufagenin, the expression of surface molecules (TLR2 and CD69) and P-p38 MAPK were also evaluated, but no significant effect was observed. Thus, our results suggest that marinobufagenin has an anti-inflammatory role in vivo and in vitro and reveals a novel possible endogenous function of this steroid in mammals.
Collapse
|
20
|
Strauss M, Smith W, Fedorova OV, Schutte AE. The Na +K +-ATPase Inhibitor Marinobufagenin and Early Cardiovascular Risk in Humans: a Review of Recent Evidence. Curr Hypertens Rep 2019; 21:38. [PMID: 30980225 PMCID: PMC6590998 DOI: 10.1007/s11906-019-0942-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW This review synthesizes recent findings in humans pertaining to the relationships between marinobufagenin (MBG), a steroidal Na+/K+-ATPase inhibitor and salt-sensitivity biomarker, and early cardiovascular risk markers. RECENT FINDINGS Twenty-four-hour urinary MBG strongly associates with habitual salt intake in young healthy adults (aged 20-30 years). Furthermore, in young healthy adults free of detected cardiovascular disease, MBG associates with increased large artery stiffness and left ventricular mass independent of blood pressure. These findings in human studies corroborate mechanistic data from rat studies whereby stimulation of MBG by a high salt intake or MBG infusion increased vascular fibrosis and cardiac hypertrophy. Twenty-four-hour urinary MBG may be a potential biomarker of early cardiovascular risk. Adverse associations between MBG-which increases with salt consumption-and early cardiovascular risk markers support the global efforts to reduce population-wide salt intake in an effort to prevent and control the burden of non-communicable diseases.
Collapse
Affiliation(s)
- Michél Strauss
- Hypertension in Africa Research Team (HART), North-West University, Private Bag X1290, Potchefstroom, 2520, South Africa
| | - Wayne Smith
- Hypertension in Africa Research Team (HART), North-West University, Private Bag X1290, Potchefstroom, 2520, South Africa
- MRC Research Unit: Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Olga V Fedorova
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Aletta E Schutte
- Hypertension in Africa Research Team (HART), North-West University, Private Bag X1290, Potchefstroom, 2520, South Africa.
- MRC Research Unit: Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa.
| |
Collapse
|
21
|
Lenaerts C, Bond L, Tuytten R, Blankert B. Revealing of endogenous Marinobufagin by an ultra-specific and sensitive UHPLC-MS/MS assay in pregnant women. Talanta 2018; 187:193-199. [PMID: 29853035 DOI: 10.1016/j.talanta.2018.05.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 05/02/2018] [Accepted: 05/04/2018] [Indexed: 01/24/2023]
Abstract
Marinobufagenin (MBG) is a bufadienolide cardiac inotrope implicated in volume expansion-mediated hypertensive states including essential hypertension and preeclampsia (PE). Endogenous MBG is an inhibitor of the α1-isoform of Na+,K+-ATPase with vasoconstrictive and cardiotonic properties, causing hypertension and natriuresis. Elevated endogenous MBG-like material levels have been described by immunoassays in salt-sensitive pregnant and preeclamptic rats as well as in preeclamptic human patients. The rise of endogenous MBG-like material appears prior the development of the main symptoms of PE, leading us to consider MBG as one of the potential biomarkers for PE. The weak specificity and the high variability of the published immunoassays gives no certification about endogenous MBG existence. This led us to set-up a highly specific and sensitive analytical method to detect MBG in plasma at low levels relying on liquid chromatography combined to mass spectrometry (UHPLC-MS/MS) with recording of 7 highly specific MRM transitions for MBG. Pure MBG standard used in the method development was obtained by purification from the Bufo marinus toad venom. d3-25-hydroxyvitamin D3 was used as internal standard. An increasing organic gradient with mobile phase A and B composed of 97:3 (v/v) H2O: MeOH and 50:45:5 (v/v/v) MeOH:IPA:H2O at pH 4.5 respectively was used on a Pursuit 3 PFP column (100 mm × 3 mm; 3 µm) to allow elution and separation of the plasmatic compounds. Chromatographic analyses of plasma samples were preceded by a precipitation of proteins pretreatment. The developed UHPLC-MS/MS assay has been applied to early-pregnant women plasma samples allowing us to investigate MBG plasma levels. Thanks to the high specificity of the assay we were able to authenticate and certify the presence of endogenous MBG in early-pregnant women plasma with the use of the 7 selected specific mass transitions. These pioneering preliminary results are giving a promising perspective for early preeclampsia risk assessment in pregnant women.
Collapse
Affiliation(s)
- Charline Lenaerts
- Laboratory of Pharmaceutical Analysis, Faculty of medicine and pharmacy, Research Institute for Health Sciences and Technology, University of Mons, Place du Parc 20, 7000 Mons, Belgium
| | - Liz Bond
- Metabolomic Diagnostics, Little Island, Cork, Ireland
| | - Robin Tuytten
- Metabolomic Diagnostics, Little Island, Cork, Ireland
| | - Bertrand Blankert
- Laboratory of Pharmaceutical Analysis, Faculty of medicine and pharmacy, Research Institute for Health Sciences and Technology, University of Mons, Place du Parc 20, 7000 Mons, Belgium.
| |
Collapse
|
22
|
Lan YL, Wang X, Lou JC, Xing JS, Zou S, Yu ZL, Ma XC, Wang H, Zhang B. Marinobufagenin inhibits glioma growth through sodium pump α1 subunit and ERK signaling-mediated mitochondrial apoptotic pathway. Cancer Med 2018; 7:2034-2047. [PMID: 29582577 PMCID: PMC5943480 DOI: 10.1002/cam4.1469] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/02/2018] [Accepted: 03/01/2018] [Indexed: 11/28/2022] Open
Abstract
Malignant glioma is one of the most challenging central nervous system diseases to treat and has high rates of recurrence and mortality. Current therapies often fail to control tumor progression or improve patient survival. Marinobufagenin (MBG) is an endogenous mammalian cardiotonic steroid involved in sodium pump inhibition. Currently, various studies have indicated the potential of MBG in cancer treatments; however, the precise mechanisms are poorly understood. The functions of MBG were examined using colony formation, migration, cell cycle, and apoptosis assays in glioma cells. A mitochondrial membrane potential assay was performed to determine the mitochondrial transmembrane potential change, and cytochrome c release from mitochondria was assayed by fluorescence microscopy. An immunofluorescence assay was performed, and the nuclear translocation of NF‐κB in glioma cells was confirmed by confocal microscopy. Western blotting and RT‐qPCR were used to detect the protein and gene expression levels, respectively. In addition, transfection experiment of ATP1A1‐siRNA was further carried out to confirm the role of sodium pump α1 subunit in the anticancer effect of MBG in human glioma. The apoptosis‐promoting and anti‐inflammatory effects of MBG were further investigated, and the sodium pump α1 subunit and the ERK signaling pathway were found to be involved in the anticancer effect of MBG. The in vivo anticancer efficacy of MBG was also tested in xenografts in nude mice. Thus, therapies targeting the ERK signaling‐mediated mitochondrial apoptotic pathways regulated by MBG might represent potential treatments for human glioma, and this study could accelerate the finding of newer therapeutic approaches for malignant glioma treatment.
Collapse
Affiliation(s)
- Yu-Long Lan
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China.,Department of Pharmacy, Dalian Medical University, Dalian, 116044, China.,Department of Physiology, Dalian Medical University, Dalian, 116044, China
| | - Xun Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Jia-Cheng Lou
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Jin-Shan Xing
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Shuang Zou
- Department of Physiology, Dalian Medical University, Dalian, 116044, China
| | - Zhen-Long Yu
- Department of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Xiao-Chi Ma
- Department of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Hongjin Wang
- Department of Neurology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Bo Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| |
Collapse
|
23
|
Strauss M, Smith W, Wei W, Fedorova OV, Schutte AE. Marinobufagenin is related to elevated central and 24-h systolic blood pressures in young black women: the African-PREDICT Study. Hypertens Res 2018; 41:183-192. [PMID: 29335615 PMCID: PMC6015745 DOI: 10.1038/s41440-017-0009-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/05/2017] [Accepted: 05/19/2017] [Indexed: 12/28/2022]
Abstract
Marinobufagenin (MBG) is an endogenous steroidal α1-Na+K+-ATPase inhibitor. Because of its role in sodium handling, MBG has been associated with both antihypertensive and prohypertensive effects in normal physiology and pathology. MBG is positively associated with blood pressure in Dahl salt-sensitive rats exhibiting a similar hypertensive phenotype to black populations, characterized by impaired urinary Na+ excretion. However, clinical studies exploring blood pressure (BP)-related effects of MBG in black populations are scant. We determined whether the MBG/Na+ ratio (assessing the effectiveness of Na+ excretion resistance to MBG) is related to systolic BP (SBP) in young black men and women, compared to whites. We included 331 apparently healthy participants (20-30 years) (42.9% black, 43.8% men) on a habitual diet. We obtained 24-h and central SBP, and 24-h urinary Na+ and MBG levels. We found no ethnic differences in MBG, Na+ or MBG/Na+. MBG excretion correlated positively with Na+ excretion in all groups and to SBP in white men and black women (p ≤ 0.011). In black women only SBP related positively to MBG/Na+ in single and multi-variable adjusted regression models: central SBP (R2 = 0.26; ß = 0.28; p = 0.039), 24-h SBP (R2 = 0.46; ß = 0.30; p = 0.011), daytime (R2 = 0.38; ß = 0.28; p = 0.023) and nighttime SBP (R2 = 0.38; ß = 0.33; p = 0.009). In contrast, inverse associations of MBG/Na+ with nighttime SBP were evident in white women (r = -0.20; p = 0.038) but lost significance after multiple adjustments (R2 = 0.36; ß = -0.13; p = 0.12). We found independent positive associations of SBP with MBG/Na+ in black women. This data supports the concept that reduced MBG-mediated Na+ excretion can contribute to adverse hemodynamics.
Collapse
Affiliation(s)
- Michél Strauss
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
| | - Wayne Smith
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
- MRC Research Unit: Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Wen Wei
- National Institute on Aging, NIH, Baltimore, MD, USA
| | | | - Aletta E Schutte
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa.
- MRC Research Unit: Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa.
| |
Collapse
|
24
|
Blaustein MP. The pump, the exchanger, and the holy spirit: origins and 40-year evolution of ideas about the ouabain-Na + pump endocrine system. Am J Physiol Cell Physiol 2017; 314:C3-C26. [PMID: 28971835 DOI: 10.1152/ajpcell.00196.2017] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Two prescient 1953 publications set the stage for the elucidation of a novel endocrine system: Schatzmann's report that cardiotonic steroids (CTSs) are all Na+ pump inhibitors, and Szent-Gyorgi's suggestion that there is an endogenous "missing screw" in heart failure that CTSs like digoxin may replace. In 1977 I postulated that an endogenous Na+ pump inhibitor acts as a natriuretic hormone and simultaneously elevates blood pressure (BP) in salt-dependent hypertension. This hypothesis was based on the idea that excess renal salt retention promoted the secretion of a CTS-like hormone that inhibits renal Na+ pumps and salt reabsorption. The hormone also inhibits arterial Na+ pumps, elevates myocyte Na+ and promotes Na/Ca exchanger-mediated Ca2+ gain. This enhances vasoconstriction and arterial tone-the hallmark of hypertension. Here I describe how those ideas led to the discovery that the CTS-like hormone is endogenous ouabain (EO), a key factor in the pathogenesis of hypertension and heart failure. Seminal observations that underlie the still-emerging picture of the EO-Na+ pump endocrine system in the physiology and pathophysiology of multiple organ systems are summarized. Milestones include: 1) cloning the Na+ pump isoforms and physiological studies of mutated pumps in mice; 2) discovery that Na+ pumps are also EO-triggered signaling molecules; 3) demonstration that ouabain, but not digoxin, is hypertensinogenic; 4) elucidation of EO's roles in kidney development and cardiovascular and renal physiology and pathophysiology; 5) discovery of "brain ouabain", a component of a novel hypothalamic neuromodulatory pathway; and 6) finding that EO and its brain receptors modulate behavior and learning.
Collapse
Affiliation(s)
- Mordecai P Blaustein
- Departments of Physiology and Medicine, University of Maryland School of Medicine , Baltimore, Maryland
| |
Collapse
|
25
|
Blaustein MP. How does pressure overload cause cardiac hypertrophy and dysfunction? High-ouabain affinity cardiac Na + pumps are crucial. Am J Physiol Heart Circ Physiol 2017; 313:H919-H930. [PMID: 28733446 PMCID: PMC5792198 DOI: 10.1152/ajpheart.00131.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/17/2022]
Abstract
Left ventricular hypertrophy is frequently observed in hypertensive patients and is believed to be due to the pressure overload and cardiomyocyte stretch. Three recent reports on mice with genetically engineered Na+ pumps, however, have demonstrated that cardiac ouabain-sensitive α2-Na+ pumps play a key role in the pathogenesis of transaortic constriction-induced hypertrophy. Hypertrophy was delayed/attenuated in mice with mutant, ouabain-resistant α2-Na+ pumps and in mice with cardiac-selective knockout or transgenic overexpression of α2-Na+ pumps. The latter, seemingly paradoxical, findings can be explained by comparing the numbers of available (ouabain-free) high-affinity (α2) ouabain-binding sites in wild-type, knockout, and transgenic hearts. Conversely, hypertrophy was accelerated in α2-ouabain-resistant (R) mice in which the normally ouabain-resistant α1-Na+ pumps were mutated to an ouabain-sensitive (S) form (α1S/Sα2R/R or "SWAP" vs. wild-type or α1R/R α2S/S mice). Furthermore, transaortic constriction-induced hypertrophy in SWAP mice was prevented/reversed by immunoneutralizing circulating endogenous ouabain (EO). These findings show that EO and its receptor, ouabain-sensitive α2, are critical factors in pressure overload-induced cardiac hypertrophy. This complements reports linking elevated plasma EO to hypertension, cardiac hypertrophy, and failure in humans and elucidates the underappreciated role of the EO-Na+ pump pathway in cardiovascular disease.
Collapse
Affiliation(s)
- Mordecai P. Blaustein
- Departments of Physiology and Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
26
|
Leenen FHH, Blaustein MP, Hamlyn JM. Update on angiotensin II: new endocrine connections between the brain, adrenal glands and the cardiovascular system. Endocr Connect 2017; 6:R131-R145. [PMID: 28855243 PMCID: PMC5613704 DOI: 10.1530/ec-17-0161] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022]
Abstract
In the brain, angiotensinergic pathways play a major role in chronic regulation of cardiovascular and electrolyte homeostasis. Increases in plasma angiotensin II (Ang II), aldosterone, [Na+] and cytokines can directly activate these pathways. Chronically, these stimuli also activate a slow neuromodulatory pathway involving local aldosterone, mineralocorticoid receptors (MRs), epithelial sodium channels and endogenous ouabain (EO). This pathway increases AT1R and NADPH oxidase subunits and maintains/further increases the activity of angiotensinergic pathways. These brain pathways not only increase the setpoint of sympathetic activity per se, but also enhance its effectiveness by increasing plasma EO and EO-dependent reprogramming of arterial and cardiac function. Blockade of any step in this slow pathway or of AT1R prevents Ang II-, aldosterone- or salt and renal injury-induced forms of hypertension. MR/AT1R activation in the CNS also contributes to the activation of sympathetic activity, the circulatory and cardiac RAAS and increase in circulating cytokines in HF post MI. Chronic central infusion of an aldosterone synthase inhibitor, MR blocker or AT1R blocker prevents a major part of the structural remodeling of the heart and the decrease in LV function post MI, indicating that MR activation in the CNS post MI depends on aldosterone, locally produced in the CNS. Thus, Ang II, aldosterone and EO are not simply circulating hormones that act on the CNS but rather they are also paracrine neurohormones, locally produced in the CNS, that exert powerful effects in key CNS pathways involved in the long-term control of sympathetic and neuro-endocrine function and cardiovascular homeostasis.
Collapse
Affiliation(s)
- Frans H H Leenen
- Brain and Heart Research GroupUniversity of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Mordecai P Blaustein
- Department of PhysiologyUniversity of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of MedicineUniversity of Maryland School of Medicine, Baltimore, Maryland, USA
| | - John M Hamlyn
- Department of PhysiologyUniversity of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
27
|
Godinho AN, Costa GT, Oliveira NO, Cardi BA, Uchoa DEA, Silveira ER, Quintas LEM, Noël FG, Fonteles MC, Carvalho KM, Santos CF, Lessa LMA, do Nascimento NRF. Effects of cardiotonic steroids on isolated perfused kidney and NHE3 activity in renal proximal tubules. Biochim Biophys Acta Gen Subj 2017; 1861:1943-1950. [PMID: 28506883 DOI: 10.1016/j.bbagen.2017.05.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 05/07/2017] [Accepted: 05/11/2017] [Indexed: 12/11/2022]
Abstract
Cardiotonic steroids (CS) are known as modulators of sodium and water homeostasis. These compounds contribute to the excretion of sodium under overload conditions due to its natriuretic property related to the inhibition of the renal Na+/K+-ATPase (NKA) pump α1 isoform. NHE3, the main route for Na+ reabsorption in the proximal tubule, depends on the Na+ gradient generated by the NKA pump. In the present study we aimed to investigate the effects of marinobufagin (MBG) and telocinobufagin (TBG) on the renal function of isolated perfused rat kidney and on the inhibition of NKA activity. Furthermore, we investigated the mechanisms for the cardiotonic steroid-mediated natriuretic effect, by evaluating and comparing the effects of bufalin (BUF), ouabain (OUA), MBG and TBG on NHE3 activity in the renal proximal tubule in vivo. TBG significantly increased GFR, UF, natriuresis and kaliuresis in isolated perfused rat kidney, and inhibits the activity of NKA at a much higher rate than MBG. By stationary microperfusion technique, the perfusion with BUF, OUA, TBG or MBG promoted an inhibitory effect on NHE3 activity, whereas BUF was the most effective agent, and demonstrated a dose-dependent response, with maximal inhibition at 50nM. Furthermore, our data showed the role of NKA-Src kinase pathway in the inhibition of NHE3 by CS. Finally, a downstream step, MEK1/2-ERK1/2 was also investigated, and, similar to Src inhibition, the MEK1/2 inhibitor (U0126) suppressed the BUF effect. Our findings indicate the involvement of NKA-SRc-Kinase-Ras-Raf-ERK1/2 pathway in the downregulation of NHE3 by cardiotonic steroids in the renal proximal tubule, promoting a reduction of proximal sodium reabsorption and natriuresis.
Collapse
Affiliation(s)
- Alana N Godinho
- Biomedical Sciences Superior Institute, State University of Ceará, Fortaleza, Brazil
| | - Graciana T Costa
- Biomedical Sciences Superior Institute, State University of Ceará, Fortaleza, Brazil
| | - Nádia O Oliveira
- Biomedical Sciences Superior Institute, State University of Ceará, Fortaleza, Brazil
| | - Bruno A Cardi
- Biomedical Sciences Superior Institute, State University of Ceará, Fortaleza, Brazil
| | | | | | - Luis Eduardo M Quintas
- Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - François G Noël
- Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Manassés C Fonteles
- Biomedical Sciences Superior Institute, State University of Ceará, Fortaleza, Brazil
| | | | - Cláudia F Santos
- Biomedical Sciences Superior Institute, State University of Ceará, Fortaleza, Brazil
| | - Lucília M A Lessa
- Biomedical Sciences Superior Institute, State University of Ceará, Fortaleza, Brazil
| | | |
Collapse
|
28
|
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
|
29
|
Involvement of the Bufadienolides in the Detection and Therapy of the Acute Respiratory Distress Syndrome. Lung 2017; 195:323-332. [PMID: 28260175 DOI: 10.1007/s00408-017-9989-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/20/2017] [Indexed: 12/12/2022]
Abstract
PURPOSE The acute respiratory distress syndrome (ARDS) represents a major challenge for clinicians as well as basic scientists. The mortality rate for ARDS has been maintained within the range of 40-52%. The authors have examined the involvement of the "cardiotonic steroids" in the pathogenesis and therapy of ARDS. We have studied the possible role of the bufadienolide, marinobufagenin (MBG), in the pathogenesis of ARDS in both a rat model of ARDS and in patients afflicted with that disorder. In addition, the potential therapeutic benefit of an antagonist of MBG, resibufogenin (RBG), in an animal model has been evaluated. METHOD A syndrome resembling human ARDS was produced in the rat by exposing the animals to 100% oxygen for 48 h. In other animals, RBG was administered to these "hyperoxic" rats, and the serum MBG was measured. In human ICU patients, urinary samples were examined for levels of MBG, and the values were compared to those obtained from other ICU patients admitted with diagnoses other than ARDS. RESULTS (1) Exposure of rats to hyperoxia produced a histologic picture which resembled that of human ARDS. (2) Serum levels of MBG in the "hyperoxic" rats substantially exceeded those obtained in animals exposed to ambient oxygen levels and were reduced to normal by RBG. (3) In ARDS patients, substantial elevations in urinary MBG were obtained compared to those in non-ARDS ICU patients. CONCLUSIONS MBG may serve as an important biomarker for the development of ARDS, and RBG may represent a preventative/therapy in this disorder.
Collapse
|
30
|
Liu J, Yan Y, Nie Y, Shapiro JI. Na/K-ATPase Signaling and Salt Sensitivity: The Role of Oxidative Stress. Antioxidants (Basel) 2017; 6:E18. [PMID: 28257114 PMCID: PMC5384181 DOI: 10.3390/antiox6010018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 02/10/2017] [Accepted: 02/22/2017] [Indexed: 02/07/2023] Open
Abstract
Other than genetic regulation of salt sensitivity of blood pressure, many factors have been shown to regulate renal sodium handling which contributes to long-term blood pressure regulation and have been extensively reviewed. Here we present our progress on the Na/K-ATPase signaling mediated sodium reabsorption in renal proximal tubules, from cardiotonic steroids-mediated to reactive oxygen species (ROS)-mediated Na/K-ATPase signaling that contributes to experimental salt sensitivity.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Yanling Yan
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Ying Nie
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Joseph I Shapiro
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| |
Collapse
|
31
|
Shah PT, Martin R, Yan Y, Shapiro JI, Liu J. Carbonylation Modification Regulates Na/K-ATPase Signaling and Salt Sensitivity: A Review and a Hypothesis. Front Physiol 2016; 7:256. [PMID: 27445847 PMCID: PMC4923243 DOI: 10.3389/fphys.2016.00256] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/11/2016] [Indexed: 01/01/2023] Open
Abstract
Na/K-ATPase signaling has been implicated in different physiological and pathophysiological conditions. Accumulating evidence indicates that oxidative stress not only regulates the Na/K-ATPase enzymatic activity, but also regulates its signaling and other functions. While cardiotonic steroids (CTS)-induced increase in reactive oxygen species (ROS) generation is an intermediate step in CTS-mediated Na/K-ATPase signaling, increase in ROS alone also stimulates Na/K-ATPase signaling. Based on literature and our observations, we hypothesize that ROS have biphasic effects on Na/K-ATPase signaling, transcellular sodium transport, and urinary sodium excretion. Oxidative modulation, in particular site specific carbonylation of the Na/K-ATPase α1 subunit, is a critical step in proximal tubular Na/K-ATPase signaling and decreased transcellular sodium transport leading to increases in urinary sodium excretion. However, once this system is overstimulated, the signaling, and associated changes in sodium excretion are blunted. This review aims to evaluate ROS-mediated carbonylation of the Na/K-ATPase, and its potential role in the regulation of pump signaling and sodium reabsorption in the renal proximal tubule (RPT).
Collapse
Affiliation(s)
- Preeya T Shah
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University Huntington, WV, USA
| | - Rebecca Martin
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University Huntington, WV, USA
| | - Yanling Yan
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University Huntington, WV, USA
| | - Joseph I Shapiro
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University Huntington, WV, USA
| | - Jiang Liu
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University Huntington, WV, USA
| |
Collapse
|
32
|
Fedorova OV, Zernetkina VI, Shilova VY, Grigorova YN, Juhasz O, Wei W, Marshall CA, Lakatta EG, Bagrov AY. Synthesis of an Endogenous Steroidal Na Pump Inhibitor Marinobufagenin, Implicated in Human Cardiovascular Diseases, Is Initiated by CYP27A1 via Bile Acid Pathway. ACTA ACUST UNITED AC 2015; 8:736-45. [PMID: 26374826 DOI: 10.1161/circgenetics.115.001217] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 08/31/2015] [Indexed: 01/06/2023]
Abstract
BACKGROUND The bioactive steroid, marinobufagenin, is an endogenous Na/K-ATPase bufadienolide inhibitor that is synthesized by adrenocortical and placental cells. Marinobufagenin binding to Na/K-ATPase initiates profibrotic cell signaling, and heightened marinobufagenin levels are implicated in the pathogenesis of hypertension, preeclampsia, and chronic kidney disease. Steroids are derived from cholesterol through the traditional steroidogenesis pathway initiated by enzyme CYP11A1, and via the acidic bile acid pathway, which is controlled by enzyme CYP27A1. The mechanism of marinobufagenin biosynthesis in mammals, however, remains unknown. METHODS AND RESULTS Here, we show that post-transcriptional silencing of the CYP27A1 gene in human trophoblast and rat adrenocortical cells reduced the expression of CYP27A1 mRNA by 70%, reduced total bile acids 2-fold, and marinobufagenin levels by 67% when compared with nontreated cells or cells transfected with nontargeting siRNA. In contrast, silencing of the CYP11A1 gene did not affect marinobufagenin production in either cell culture, but suppressed production of progesterone 2-fold in human trophoblast cells and of corticosterone by 90% in rat adrenocortical cells when compared with cells transfected with nontargeting siRNA. In vivo, in a high-salt administration experiment, male and female Dahl salt-sensitive rats became hypertensive after 4 weeks on a high-NaCl diet, their plasma marinobufagenin levels doubled, and adrenocortical CYP27A1 mRNA and protein increased 1.6-fold and 2.0-fold. CONCLUSIONS Therefore, the endogenous steroidal Na/K-ATPase inhibitor, marinobufagenin, is synthesized in mammalian placenta and adrenal cortex from cholesterol through the novel acidic bile acid pathway. These findings will help to understand the role of marinobufagenin in highly prevalent human cardiovascular diseases.
Collapse
Affiliation(s)
- Olga V Fedorova
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD.
| | - Valentina I Zernetkina
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Victoria Y Shilova
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Yulia N Grigorova
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Ondrej Juhasz
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Wen Wei
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Courtney A Marshall
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Edward G Lakatta
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Alexei Y Bagrov
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD.
| |
Collapse
|
33
|
Lin S, Leonard D, Co MAM, Mukhopadhyay D, Giri B, Perger L, Beeram MR, Kuehl TJ, Uddin MN. Pre-eclampsia has an adverse impact on maternal and fetal health. Transl Res 2015; 165:449-63. [PMID: 25468481 DOI: 10.1016/j.trsl.2014.10.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/08/2014] [Accepted: 10/09/2014] [Indexed: 12/15/2022]
Abstract
Pre-eclampsia (preE) is a multifaceted complication found uniquely in the pregnant patient and one that has puzzled scientists for years. PreE is not a single disorder, but a complex syndrome that is produced by various pathophysiological triggers and mechanisms affecting about 5% of obstetrical patients. PreE is a major cause of premature delivery and maternal and fetal morbidity and mortality. PreE is characterized by de novo development of hypertension and proteinuria after 20 weeks of gestation and affects nearly every organ system, with the most severe consequences being eclampsia, pulmonary edema, intrauterine growth restriction, and thrombocytopenia. PreE alters the intrauterine environment by modulating the pattern of hormonal signals and activating the detrimental cellular signaling that has been transported to the fetus. The fetus has to adapt to this intrauterine environment with detrimental signals. The adaptive changes increase the risk of disease later in life. This review defines the predisposition and causes of preE and the cellular signaling detrimental to maternal health during preE. Moreover, the risk factors for diseases that are transmitted to the offspring have been addressed in this review. The detrimental signaling molecules that have been overexpressed in preE patients raises the possibility that those signals could be therapeutically blocked one day.
Collapse
Affiliation(s)
- Saunders Lin
- Texas A&M University College of Medicine, Temple, Tex
| | | | - Mary A M Co
- Department of Pediatrics, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex
| | - Dhriti Mukhopadhyay
- Department of Surgery, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex
| | - Badri Giri
- Department of Pulmonary Critical Care, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex
| | - Lena Perger
- Department of Surgery, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex
| | - Madhava R Beeram
- Texas A&M University College of Medicine, Temple, Tex; Department of Pediatrics, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex
| | - Thomas J Kuehl
- Texas A&M University College of Medicine, Temple, Tex; Department of Pediatrics, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex; Department of Obstetrics and Gynecology, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex
| | - Mohammad N Uddin
- Texas A&M University College of Medicine, Temple, Tex; Prehealth Studies, Baylor University, Waco, Tex; Department of Pediatrics, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex; Department of Obstetrics and Gynecology, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex.
| |
Collapse
|
34
|
Ning J, Yu ZL, Hu LH, Wang C, Huo XK, Deng S, Hou J, Wu JJ, Ge GB, Ma XC, Yang L. Characterization of phase I metabolism of resibufogenin and evaluation of the metabolic effects on its antitumor activity and toxicity. Drug Metab Dispos 2015; 43:299-308. [PMID: 25504504 DOI: 10.1124/dmd.114.060996] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Resibufogenin (RB), one of the major active compounds of the traditional Chinese medicine Chansu, has displayed great potential as a chemotherapeutic agent in oncology. However, it is a digoxin-like compound that also exhibits extremely cardiotoxic effects. The present study aimed to characterize the metabolic behaviors of RB in humans as well as to evaluate the metabolic effects on its bioactivity and toxicity. The phase I metabolic profile in human liver microsomes was characterized systemically, and the major metabolite was identified as marinobufagenin (5β-hydroxylresibufogenin, 5-HRB) by liquid chromatography-mass spectrometry and nuclear magnetic imaging techniques. Both cytochrome P450 (P450) reaction phenotyping and inhibition assays using P450-selective chemical inhibitors demonstrated that CYP3A4 was mainly involved in RB 5β-hydroxylation with much higher selectivity than CYP3A5. Kinetic characterization demonstrated that RB 5β-hydroxylation in both human liver microsomes and human recombinant CYP3A4 obeyed biphasic kinetics and displayed similar apparent kinetic parameters. Furthermore, 5-HRB could significantly induce cell growth inhibition and apoptosis in A549 and H1299 by facilitating apoptosome assembly and caspase activation. Meanwhile, 5-HRB displayed very weak cytotoxicity of human embryonic lung fibroblasts, and in mice there was a greater tolerance to acute toxicity. In summary, CYP3A4 dominantly mediated 5β-hydroxylation and was found to be a major metabolic pathway of RB in the human liver, whereas its major metabolite (5-HRB) displayed better druglikeness than its parent compound RB. Our findings lay a solid foundation for RB metabolism studies in humans and encourage further research on the bioactive metabolite of RB.
Collapse
Affiliation(s)
- Jing Ning
- Research Institute of Integrated Traditional and Western Medicine, College of Pharmacy, Dalian Medical University, Dalian, China (J.N., Z.-L.Y., C.W., X.-K H., S.D., J.H., X.-C.M.); Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, Jilin University, Changchun, China (L.-H., H); Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (J.N., J.-J.W., G.-B.G., L.Y.); State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (C.W.)
| | - Zhen-Long Yu
- Research Institute of Integrated Traditional and Western Medicine, College of Pharmacy, Dalian Medical University, Dalian, China (J.N., Z.-L.Y., C.W., X.-K H., S.D., J.H., X.-C.M.); Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, Jilin University, Changchun, China (L.-H., H); Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (J.N., J.-J.W., G.-B.G., L.Y.); State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (C.W.)
| | - Liang-Hai Hu
- Research Institute of Integrated Traditional and Western Medicine, College of Pharmacy, Dalian Medical University, Dalian, China (J.N., Z.-L.Y., C.W., X.-K H., S.D., J.H., X.-C.M.); Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, Jilin University, Changchun, China (L.-H., H); Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (J.N., J.-J.W., G.-B.G., L.Y.); State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (C.W.)
| | - Chao Wang
- Research Institute of Integrated Traditional and Western Medicine, College of Pharmacy, Dalian Medical University, Dalian, China (J.N., Z.-L.Y., C.W., X.-K H., S.D., J.H., X.-C.M.); Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, Jilin University, Changchun, China (L.-H., H); Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (J.N., J.-J.W., G.-B.G., L.Y.); State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (C.W.)
| | - Xiao-Kui Huo
- Research Institute of Integrated Traditional and Western Medicine, College of Pharmacy, Dalian Medical University, Dalian, China (J.N., Z.-L.Y., C.W., X.-K H., S.D., J.H., X.-C.M.); Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, Jilin University, Changchun, China (L.-H., H); Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (J.N., J.-J.W., G.-B.G., L.Y.); State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (C.W.)
| | - Sa Deng
- Research Institute of Integrated Traditional and Western Medicine, College of Pharmacy, Dalian Medical University, Dalian, China (J.N., Z.-L.Y., C.W., X.-K H., S.D., J.H., X.-C.M.); Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, Jilin University, Changchun, China (L.-H., H); Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (J.N., J.-J.W., G.-B.G., L.Y.); State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (C.W.)
| | - Jie Hou
- Research Institute of Integrated Traditional and Western Medicine, College of Pharmacy, Dalian Medical University, Dalian, China (J.N., Z.-L.Y., C.W., X.-K H., S.D., J.H., X.-C.M.); Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, Jilin University, Changchun, China (L.-H., H); Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (J.N., J.-J.W., G.-B.G., L.Y.); State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (C.W.)
| | - Jing-Jing Wu
- Research Institute of Integrated Traditional and Western Medicine, College of Pharmacy, Dalian Medical University, Dalian, China (J.N., Z.-L.Y., C.W., X.-K H., S.D., J.H., X.-C.M.); Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, Jilin University, Changchun, China (L.-H., H); Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (J.N., J.-J.W., G.-B.G., L.Y.); State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (C.W.)
| | - Guang-Bo Ge
- Research Institute of Integrated Traditional and Western Medicine, College of Pharmacy, Dalian Medical University, Dalian, China (J.N., Z.-L.Y., C.W., X.-K H., S.D., J.H., X.-C.M.); Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, Jilin University, Changchun, China (L.-H., H); Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (J.N., J.-J.W., G.-B.G., L.Y.); State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (C.W.)
| | - Xiao-Chi Ma
- Research Institute of Integrated Traditional and Western Medicine, College of Pharmacy, Dalian Medical University, Dalian, China (J.N., Z.-L.Y., C.W., X.-K H., S.D., J.H., X.-C.M.); Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, Jilin University, Changchun, China (L.-H., H); Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (J.N., J.-J.W., G.-B.G., L.Y.); State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (C.W.)
| | - Ling Yang
- Research Institute of Integrated Traditional and Western Medicine, College of Pharmacy, Dalian Medical University, Dalian, China (J.N., Z.-L.Y., C.W., X.-K H., S.D., J.H., X.-C.M.); Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, Jilin University, Changchun, China (L.-H., H); Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China (J.N., J.-J.W., G.-B.G., L.Y.); State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (C.W.)
| |
Collapse
|
35
|
Pavlovic D. The role of cardiotonic steroids in the pathogenesis of cardiomyopathy in chronic kidney disease. Nephron Clin Pract 2014; 128:11-21. [PMID: 25341357 DOI: 10.1159/000363301] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Cardiotonic steroids (CTS) are a new class of hormones that circulate in the blood and are divided into two distinct groups, cardenolides, such as ouabain and digoxin, and bufadienolides, such as marinobufagenin, telocinobufagin and bufalin. They have the ability to bind and inhibit the ubiquitous transport enzyme sodium potassium pump, thus regulating intracellular Na(+) concentration in every living cell. Although digoxin has been prescribed to heart failure patients for at least 200 years, the realization that CTS are endogenously produced has intensified research into their physiological and pathophysiological roles. Over the last two decades, substantial evidence has accumulated demonstrating the effects of endogenously synthesised CTS on the kidneys, vasculature and the heart. In this review, the current state of art and the controversies surrounding the manner in which CTS mediate their pathophysiological effects are discussed. Several potential therapeutic strategies have emerged as a result of our increased understanding of the role CTS play in health and disease.
Collapse
Affiliation(s)
- Davor Pavlovic
- Cardiovascular Division, King's College London, Rayne Institute, St. Thomas' Hospital, London, UK
| |
Collapse
|
36
|
Jablonski KL, Fedorova OV, Racine ML, Geolfos CJ, Gates PE, Chonchol M, Fleenor BS, Lakatta EG, Bagrov AY, Seals DR. Dietary sodium restriction and association with urinary marinobufagenin, blood pressure, and aortic stiffness. Clin J Am Soc Nephrol 2013; 8:1952-9. [PMID: 23929930 DOI: 10.2215/cjn.00900113] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND AND OBJECTIVES Systolic BP and large elastic artery stiffness both increase with age and are reduced by dietary sodium restriction. Production of the natriuretic hormone marinobufagenin, an endogenous α1 Na+,K+-ATPase inhibitor, is increased in salt-sensitive hypertension and contributes to the rise in systolic BP during sodium loading. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS The hypothesis was that dietary sodium restriction performed in middle-aged/older adults (eight men and three women; 60 ± 2 years) with moderately elevated systolic BP (139 ± 2/83 ± 2 mmHg) would reduce urinary marinobufagenin excretion as well as systolic BP and aortic pulse-wave velocity (randomized, placebo-controlled, and crossover design). This study also explored the associations among marinobufagenin excretion with systolic BP and aortic pulse-wave velocity across conditions of 5 weeks of a low-sodium (77 ± 9 mmol/d) and 5 weeks of a normal-sodium (144 ± 7 mmol/d) diet. RESULTS Urinary marinobufagenin excretion (weekly measurements; 25.4 ± 1.8 versus 30.7 ± 2.1 pmol/kg per day), systolic BP (127 ± 3 versus 138 ± 5 mmHg), and aortic pulse-wave velocity (700 ± 40 versus 843 ± 36 cm/s) were lower during the low- versus normal-sodium condition (all P<0.05). Across all weeks, marinobufagenin excretion was related with systolic BP (slope=0.61, P<0.001) and sodium excretion (slope=0.46, P<0.001). These associations persisted during the normal- but not the low-sodium condition (both P<0.005). Marinobufagenin excretion also was associated with aortic pulse-wave velocity (slope=0.70, P=0.02) and endothelial cell expression of NAD(P)H oxidase-p47phox (slope=0.64, P=0.006). CONCLUSIONS These results show, for the first time in humans, that dietary sodium restriction reduces urinary marinobufagenin excretion and that urinary marinobufagenin excretion is positively associated with systolic BP, aortic stiffness (aortic pulse-wave velocity), and endothelial cell expression of the oxidant enzyme NAD(P)H oxidase. Importantly, marinobufagenin excretion is positively related to systolic BP over ranges of sodium intake typical of an American diet, extending previous observations in rodents and humans fed experimentally high-sodium diets.
Collapse
Affiliation(s)
- Kristen L Jablonski
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado;, †Intramural Research Program, Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland;, ‡Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom, §Division of Renal Diseases and Hypertension, University of Colorado Denver Anschutz Medical Center, Aurora, Colorado
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Zicha J, Dobešová Z, Vokurková M, Rauchová H, Hojná S, Kadlecová M, Behuliak M, Vaněčková I, Kuneš J. Age-dependent salt hypertension in Dahl rats: fifty years of research. Physiol Res 2013; 61:S35-S87. [PMID: 22827876 DOI: 10.33549/physiolres.932363] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Fifty years ago, Lewis K. Dahl has presented a new model of salt hypertension - salt-sensitive and salt-resistant Dahl rats. Twenty years later, John P. Rapp has published the first and so far the only comprehensive review on this rat model covering numerous aspects of pathophysiology and genetics of salt hypertension. When we summarized 25 years of our own research on Dahl/Rapp rats, we have realized the need to outline principal abnormalities of this model, to show their interactions at different levels of the organism and to highlight the ontogenetic aspects of salt hypertension development. Our attention was focused on some cellular aspects (cell membrane function, ion transport, cell calcium handling), intra- and extrarenal factors affecting renal function and/or renal injury, local and systemic effects of renin-angiotensin-aldosterone system, endothelial and smooth muscle changes responsible for abnormal vascular contraction or relaxation, altered balance between various vasoconstrictor and vasodilator systems in blood pressure maintenance as well as on the central nervous and peripheral mechanisms involved in the regulation of circulatory homeostasis. We also searched for the age-dependent impact of environmental and pharmacological interventions, which modify the development of high blood pressure and/or organ damage, if they influence the salt-sensitive organism in particular critical periods of development (developmental windows). Thus, severe self-sustaining salt hypertension in young Dahl rats is characterized by pronounced dysbalance between augmented sympathetic hyperactivity and relative nitric oxide deficiency, attenuated baroreflex as well as by a major increase of residual blood pressure indicating profound remodeling of resistance vessels. Salt hypertension development in young but not in adult Dahl rats can be attenuated by preventive increase of potassium or calcium intake. On the contrary, moderate salt hypertension in adult Dahl rats is attenuated by superoxide scavenging or endothelin-A receptor blockade which do not affect salt hypertension development in young animals.
Collapse
Affiliation(s)
- J Zicha
- Centre for Cardiovascular Research, Prague, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Lenaerts C, Demeyer M, Gerbaux P, Blankert B. Analytical aspects of marinobufagenin. Clin Chim Acta 2013; 421:193-201. [DOI: 10.1016/j.cca.2013.02.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 02/21/2013] [Accepted: 02/22/2013] [Indexed: 11/30/2022]
|
39
|
Uddin MN, Allen SR, Jones RO, Zawieja DC, Kuehl TJ. Pathogenesis of pre-eclampsia: marinobufagenin and angiogenic imbalance as biomarkers of the syndrome. Transl Res 2012; 160:99-113. [PMID: 22683369 DOI: 10.1016/j.trsl.2012.01.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 01/03/2012] [Accepted: 01/05/2012] [Indexed: 11/17/2022]
Abstract
Pre-eclampsia (preE), a pregnancy disorder with the de novo onset of hypertension and proteinuria after 20 weeks of gestation, has multiple triggers that initiate pathophysiologic mechanisms. This review addresses translational aspects of preE by synthesizing information on preE pathogenesis, describing diagnostic biomarkers that predict disease, and suggesting strategies to lessen adverse outcomes. Key to this understanding is the role of cardiotonic bufodienolides, with marinobufagenin (MBG) as the prototype, and angiogenic factors in preE pathogenesis. Data from a rat model believed to mimic human preE show that urinary excretion of MBG increases before the onset of hypertension and proteinuria and that affected animals have an increased vascular leakage and blood brain barrier permeability. Angiogenic imbalance occurs with the onset of the syndrome in this model. Also, we report that MBG levels in preE patients exceed those in normal pregnancy and that angiogenic factors are altered in patients showing signs and symptoms of overt disease. In vitro administration of MBG inhibits cytotrophoblast function and triggers hyperpermeability in endothelial cell monolayers. We advance the hypotheses that MBG precedes preE; MBG causes disruption of tight junction proteins leading to vascular leak via activation of MAPK which triggers apoptotic mechanisms resulting in further endothelial dysfunction leading to edema with the release of angiogenic factors. This review provides new evidence about the role of MBG and vasoactive intermediates in preE pathogenesis including the neurologic sequela and may reveal new therapeutic targets for the prevention of preE complications.
Collapse
Affiliation(s)
- Mohammad N Uddin
- Department of Obstetrics and Gynecology, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, TX 76508, USA.
| | | | | | | | | |
Collapse
|
40
|
DOCA-salt hypertension does not require the ouabain-sensitive binding site of the α2 Na,K-ATPase. Am J Hypertens 2012; 25:421-9. [PMID: 22258333 DOI: 10.1038/ajh.2011.255] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND We have shown that the ouabain-sensitive α2 Na,K-ATPase is required for adrenocorticotropic hormone (ACTH)-induced hypertension and gestational blood pressure regulation. It is therefore of interest to explore whether this binding site participates in the development of other forms of hypertension, such as deoxycorticosterone acetate (DOCA)-salt using mutant mice with altered sensitivity to ouabain. METHODS Wild-type (α1 ouabain-resistant, α2 ouabain-sensitive: α(R/R)α2(S/S)), α1-resistant, α2-resistant (α1(R/R)α2(R/R)) and α1-sensitive, α2-resistant (α1(S/S)α2(R/R)) mice were uninephrectomized and implanted with DOCA pellets. The animals were given either tap water or 1% NaCl, and blood pressure was measured before and after DOCA. RESULTS DOCA-salt-treated α1(R/R)α2(R/R) mice developed hypertension to the same extent as α1(R/R)α2(S/S) mice (wild type), and the α1(S/S)α2(R/R) mice given DOCA-salt also showed no difference from the other two genotypes. The expression of the α1 isoform was not changed by DOCA-salt treatment in either α1(R/R)α2(S/S) or α1(R/R)α2(R/R) mice. However, the α2 subunit was expressed at substantially higher levels in the hearts of α1(R/R)α2(R/R) than α1(R/R)α2(S/S) mice, regardless of treatment. Plasma levels of ouabain did not change consistently, but those of marinobufagenin were modestly higher in DOCA-salt treated mice relatively to those without salt. CONCLUSIONS The ouabain-binding site of either the α1 or α2 Na,K-ATPase subunit does not play an essential role in the development of DOCA-salt hypertension in this mouse model. These findings indicate that the underlying mechanisms of hypertension induced by DOCA-salt treatment are different from those of ACTH-induced hypertension.
Collapse
|
41
|
Reactive Oxygen Species Modulation of Na/K-ATPase Regulates Fibrosis and Renal Proximal Tubular Sodium Handling. Int J Nephrol 2012; 2012:381320. [PMID: 22518311 PMCID: PMC3299271 DOI: 10.1155/2012/381320] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 11/07/2011] [Indexed: 01/11/2023] Open
Abstract
The Na/K-ATPase is the primary force regulating renal sodium handling and plays a key role in both ion homeostasis and blood pressure regulation. Recently, cardiotonic steroids (CTS)-mediated Na/K-ATPase signaling has been shown to regulate fibrosis, renal proximal tubule (RPT) sodium reabsorption, and experimental Dahl salt-sensitive hypertension in response to a high-salt diet. Reactive oxygen species (ROS) are an important modulator of nephron ion transport. As there is limited knowledge regarding the role of ROS-mediated fibrosis and RPT sodium reabsorption through the Na/K-ATPase, the focus of this review is to examine the possible role of ROS in the regulation of Na/K-ATPase activity, its signaling, fibrosis, and RPT sodium reabsorption.
Collapse
|
42
|
Takahashi H, Yoshika M, Komiyama Y, Nishimura M. The central mechanism underlying hypertension: a review of the roles of sodium ions, epithelial sodium channels, the renin-angiotensin-aldosterone system, oxidative stress and endogenous digitalis in the brain. Hypertens Res 2011; 34:1147-60. [PMID: 21814209 PMCID: PMC3324327 DOI: 10.1038/hr.2011.105] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 05/08/2011] [Accepted: 05/15/2011] [Indexed: 02/07/2023]
Abstract
The central nervous system has a key role in regulating the circulatory system by modulating the sympathetic and parasympathetic nervous systems, pituitary hormone release, and the baroreceptor reflex. Digoxin- and ouabain-like immunoreactive materials were found >20 years ago in the hypothalamic nuclei. These factors appeared to localize to the paraventricular and supraoptic nuclei and the nerve fibers at the circumventricular organs and supposed to affect electrolyte balance and blood pressure. The turnover rate of these materials increases with increasing sodium intake. As intracerebroventricular injection of ouabain increases blood pressure via sympathetic activation, an endogenous digitalis-like factor (EDLF) was thought to regulate cardiovascular system-related functions in the brain, particularly after sodium loading. Experiments conducted mainly in rats revealed that the mechanism of action of ouabain in the brain involves sodium ions, epithelial sodium channels (ENaCs) and the renin-angiotensin-aldosterone system (RAAS), all of which are affected by sodium loading. Rats fed a high-sodium diet develop elevated sodium levels in their cerebrospinal fluid, which activates ENaCs. Activated ENaCs and/or increased intracellular sodium in neurons activate the RAAS; this releases EDLF in the brain, activating the sympathetic nervous system. The RAAS promotes oxidative stress in the brain, further activating the RAAS and augmenting sympathetic outflow. Angiotensin II and aldosterone of peripheral origin act in the brain to activate this cascade, increasing sympathetic outflow and leading to hypertension. Thus, the brain Na(+)-ENaC-RAAS-EDLF axis activates sympathetic outflow and has a crucial role in essential and secondary hypertension. This report provides an overview of the central mechanism underlying hypertension and discusses the use of antihypertensive agents.
Collapse
Affiliation(s)
- Hakuo Takahashi
- Department of Clinical Sciences and Laboratory Medicine, Kansai Medical University, Hirakata City, Osaka, Japan.
| | | | | | | |
Collapse
|
43
|
Koleganova N, Piecha G, Ritz E, Becker LE, Müller A, Weckbach M, Nyengaard JR, Schirmacher P, Gross-Weissmann ML. Both high and low maternal salt intake in pregnancy alter kidney development in the offspring. Am J Physiol Renal Physiol 2011; 301:F344-54. [PMID: 21593188 DOI: 10.1152/ajprenal.00626.2010] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In humans, low glomerular numbers are related to hypertension, cardiovascular, and renal disease in adult life. The present study was designed 1) to explore whether above- or below-normal dietary salt intake during pregnancy influences nephron number and blood pressure in the offspring and 2) to identify potential mechanisms in kidney development modified by maternal sodium intake. Sprague-Dawley rats were fed low (0.07%)-, intermediate (0.51%)-, or high (3.0%)-sodium diets during pregnancy and lactation. The offspring were weaned at 4 wk and subsequently kept on a 0.51% sodium diet. The kidney structure was assessed at postnatal weeks 1 and 12 and the expression of proteins of interest at term and at week 1. Blood pressure was measured in male offspring by telemetry from postnatal month 2 to postnatal month 9. The numbers of glomeruli at weeks 1 and 12 were significantly lower and, in males, telemetrically measured mean arterial blood pressure after month 5 was higher in offspring of dams on a high- or low- compared with intermediate-sodium diet. A high-salt diet was paralleled by higher concentrations of marinobufagenin in the amniotic fluid and an increase in the expression of both sprouty-1 and glial cell-derived neutrophic factor in the offspring's kidney. The expression of FGF-10 was lower in offspring of dams on a low-sodium diet, and the expression of Pax-2 and FGF-2 was lower in offspring of dams on a high-sodium diet. Both excessively high and excessively low sodium intakes during pregnancy modify protein expression in offspring kidneys and reduce the final number of glomeruli, predisposing the risk of hypertension later in life.
Collapse
|
44
|
Kolmakova EV, Haller ST, Kennedy DJ, Isachkina AN, Budny GV, Frolova EV, Piecha G, Nikitina ER, Malhotra D, Fedorova OV, Shapiro JI, Bagrov AY. Endogenous cardiotonic steroids in chronic renal failure. Nephrol Dial Transplant 2011; 26:2912-9. [PMID: 21292813 DOI: 10.1093/ndt/gfq772] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Previous reports demonstrated that digitalis-like cardiotonic steroids (CTS) contribute to the pathogenesis of end-stage renal disease. The goal of the present study was to define the nature of CTS in patients with chronic kidney disease (CKD) and in partially nephrectomized (PNx) rats. METHODS In patients with CKD and in healthy controls, we determined plasma levels of marinobufagenin (MBG) and endogenous ouabain (EO) and erythrocyte Na/K-ATPase activity in the absence and in the presence of 3E9 anti-MBG monoclonal antibody (mAb) and Digibind. Levels of MBG and EO were also determined in sham-operated Sprague-Dawley rats and in rats following 4 weeks of PNx. RESULTS In 25 patients with CKD plasma, MBG but not EO was increased (0.86 ± 0.07 versus 0.28 ± 0.02 nmol/L, P < 0.01) and erythrocyte Na/K-ATPase was inhibited (1.24 ± 0.10 versus 2.80 ± 0.09 μmol Pi/mL/h, P < 0.01) as compared to that in 19 healthy subjects. Ex vivo, 3E9 mAb restored Na/K-ATPase in erythrocytes from patients with CKD but did not affect Na/K-ATPase from control subjects. Following chromatographic fractionation of uremic versus normal plasma, a competitive immunoassay based on anti-MBG mAb detected a 3-fold increase in the level of endogenous material having retention time similar to that seen with MBG. A similar pattern of CTS changes was observed in uremic rats. As compared to sham-operated animals, PNx rats exhibited 3-fold elevated levels of MBG but not that of EO. CONCLUSIONS In chronic renal failure, elevated levels of a bufadienolide CTS, MBG, contribute to Na/K-ATPase inhibition and may represent a potential target for therapy.
Collapse
Affiliation(s)
- Elena V Kolmakova
- Chronic Dialysis Division, Mechnikov Medical Academy, St. Petersburg, Russia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Gao H, Popescu R, Kopp B, Wang Z. Bufadienolides and their antitumor activity. Nat Prod Rep 2011; 28:953-69. [DOI: 10.1039/c0np00032a] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
46
|
Fedorova OV, Shapiro JI, Bagrov AY. Endogenous cardiotonic steroids and salt-sensitive hypertension. BIOCHIMICA ET BIOPHYSICA ACTA 2010; 1802:1230-6. [PMID: 20347967 PMCID: PMC2933321 DOI: 10.1016/j.bbadis.2010.03.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Revised: 03/18/2010] [Accepted: 03/20/2010] [Indexed: 12/12/2022]
Abstract
Endogenous cardiotonic steroids (CTS), also called digitalis like factors, have been postulated to play important roles in pathogenesis of hypertension for nearly half of a century. For the past 50 years biomedical scientists have been in quest of an unidentified factor or hormone that both increases blood pressure and renal sodium excretion; this "natriuretic hormone" was, in fact, postulated to interact with the Na/K-ATPase. Recent discoveries have led to the identification of steroid molecules which are present in humans, rodents and amphibians, and which, in a complex manner, interact with each other and with the other systems that regulate renal salt handling and contribute to the salt-sensitivity of blood pressure. Recent findings include the specific identification of endogenous cardenolide (endogenous ouabain) and bufadienolide (marinobufagenin) CTS in humans along with the delineation of mechanisms by which CTS can signal through the Na/K-ATPase. Although CTS were first considered important in the regulation of renal sodium transport and arterial pressure, more recent work implicates these hormones in the central regulation of blood pressure and regulation of cell growth, and development of cardiovascular and renal fibrosis in particular.
Collapse
Affiliation(s)
- Olga V. Fedorova
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | | | - Alexei Y. Bagrov
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD
| |
Collapse
|
47
|
Liu J, Xie ZJ. The sodium pump and cardiotonic steroids-induced signal transduction protein kinases and calcium-signaling microdomain in regulation of transporter trafficking. BIOCHIMICA ET BIOPHYSICA ACTA 2010; 1802:1237-45. [PMID: 20144708 PMCID: PMC5375027 DOI: 10.1016/j.bbadis.2010.01.013] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 01/28/2010] [Accepted: 01/30/2010] [Indexed: 12/12/2022]
Abstract
The Na/K-ATPase was discovered as an energy transducing ion pump. A major difference between the Na/K-ATPase and other P-type ATPases is its ability to bind a group of chemicals called cardiotonic steroids (CTS). The plant-derived CTS such as digoxin are valuable drugs for the management of cardiac diseases, whereas ouabain and marinobufagenin (MBG) have been identified as a new class of endogenous hormones. Recent studies have demonstrated that the endogenous CTS are important regulators of renal Na(+) excretion and blood pressure. The Na/K-ATPase is not only an ion pump, but also an important receptor that can transduce the ligand-like effect of CTS on intracellular protein kinases and Ca(2+) signaling. Significantly, these CTS-provoked signaling events are capable of reducing the surface expression of apical NHE3 (Na/H exchanger isoform 3) and basolateral Na/K-ATPase in renal proximal tubular cells. These findings suggest that endogenous CTS may play an important role in regulation of tubular Na(+) excretion under physiological conditions; conversely, a defect at either the receptor level (Na/K-ATPase) or receptor-effector coupling would reduce the ability of renal proximal tubular cells to excrete Na(+), thus culminating/resulting in salt-sensitive hypertension.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Medicine, University of Toledo College of Medicine
| | - Zi-jian Xie
- Department of Physiology and Pharmacology, University of Toledo College of Medicine
| |
Collapse
|
48
|
Abstract
Current salt intake is too high. Current evidence documents that salt is crucial to the genesis of hypertension.
Collapse
Affiliation(s)
- Eberhard Ritz
- Department of Internal Medicine, Division Nephrology, Nierenzentrum, Heidelberg, Germany.
| |
Collapse
|
49
|
Uddin MN, Horvat D, Demorrow S, Agunanne E, Puschett JB. Marinobufagenin is an upstream modulator of Gadd45a stress signaling in preeclampsia. Biochim Biophys Acta Mol Basis Dis 2010; 1812:49-58. [PMID: 20851181 DOI: 10.1016/j.bbadis.2010.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Revised: 09/09/2010] [Accepted: 09/13/2010] [Indexed: 11/18/2022]
Abstract
Preeclampsia (PE) is a hypertensive disorder of pregnancy, in which marinobufagenin (MBG), a circulating cardiotonic steroid, is increased. The Gadd45a stress sensor protein is an upstream modulator of the pathophysiological changes observed in PE. However, the effects of MBG on Gadd45a stress signaling remain unknown. We examined the expression of Gadd45a, the sFlt-1 receptor, and p38, as well as caspase 3 and 8 activities in placental samples from four groups of rats. These were: normal pregnant (NP, n=8); pregnant rats which received weekly injections of desoxycorticosterone acetate and 0.9% saline as their drinking water (PDS, n=9); normal pregnant rats injected with MBG (NPM, n=8); and PDS rats injected with resibufogenin (RBG), an in vivo antagonist of MBG (PDSR, n=8). Utilizing human cytotrophoblast (CTB) cells, we examined the effect of MBG on these stress signaling proteins in vitro. Placental Gadd45a expression, caspase 3 and 8 activities, sFlt-1 concentrations, and sFlt-1 receptor expression were significantly higher in PDS and NPM compared to NP and PDSR rats. Gadd45a protein was significantly upregulated in the CTB cells when MBG was present in concentrations ≥1nM. Treatment with MBG (≥1nM) also significantly arrested cell cycle progression and activated the expression of the Gadd45a-mediated stress signaling proteins. Inhibition of Gadd45a through RNAi-mediation attenuated MBG-induced CTB cell stress signaling. In conclusion, MBG is involved in the alteration in Gadd45a stress signaling both in vivo and in vitro and RBG prevents these changes when administered in vivo.
Collapse
Affiliation(s)
- Mohammad N Uddin
- Division of Nephrology and Hypertension, Department of Medicine, Texas A&M Health Science Center College of Medicine and Scott & White Memorial Hospital, Temple, TX 76508, USA
| | | | | | | | | |
Collapse
|
50
|
Puschett JB, Agunanne E, Uddin MN. Emerging Role of the Bufadienolides in Cardiovascular and Kidney Diseases. Am J Kidney Dis 2010; 56:359-70. [DOI: 10.1053/j.ajkd.2010.01.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Accepted: 01/05/2010] [Indexed: 01/11/2023]
|