1
|
Poluektov YM, Lopina OD, Strelkova MA, Kuleshova ID, Makarov AA, Petrushanko IY. Mechanisms mediating effects of cardiotonic steroids in mammalian blood cells. Front Pharmacol 2025; 16:1520927. [PMID: 40196366 PMCID: PMC11973394 DOI: 10.3389/fphar.2025.1520927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/21/2025] [Indexed: 04/09/2025] Open
Abstract
Cardiotonic steroids (CTSs) were known as steroidal plant compounds that exert cellular effects by the binding to Na,K-ATPase. Earlier, plant (exogenous) CTSs were used to treat chronic heart failure. By now, endogenous CTS have been identified in mammals, and their concentrations in the blood, normally in a subnanomolar range, are altered in numerous pathologies. This indicates their role as endogenous regulators of physiological processes. CTS transport occurs primarily in the blood, yet the CTS effects on blood cells remain poorly understood. This review summarizes the CTS effects on blood cells of animals and humans under normal and pathological conditions, and analyzes their action based on known mechanisms of action in mammalian cells. At high concentrations (greater than 10-9 M), CTS binding to Na,K-ATPase inhibits the enzyme, whereas lower concentrations of CTSs induce signaling cascades or activate the enzyme. All these mechanisms are shown to be present in blood cells. The particular CTS effect is determined by the CTS type, its concentration, the isoform composition of the catalytic α-subunit of Na,K-ATPase in the cell, and other cell features. It has been demonstrated that all blood cell types (erythrocytes, leukocytes, and platelets) expressed both ubiquitously distributed α1-isoform and tissue-specific α3-subunit, which exhibits a different ion and CTS affinity compared to α1. This results in a wide spectrum of blood cell responses to fluctuations in CTS levels in the blood. In particular, an increase in the level of endogenous CTSs by a more twofold is sufficient to induce a decline in the activity of erythrocyte Na,K-ATPase. The administration of exogenous CTSs is able to modulate the proinflammatory activity of leukocytes, which is attributed to the activation of signaling cascades, and to exert an influence on platelet activation. Hence, alterations of CTS levels in bloodstream significantly affect the functionality of blood cells, contributing to the organism's adaptive response. On top of this, a comparison of the effects of CTSs on human leukocytes and rodent leukocytes carrying the CTS-resistant α1-isoform often reveals opposite effects, thus indicating that rodents are an unsuitable model for studying CTS effects on these cells.
Collapse
Affiliation(s)
- Yuri M. Poluektov
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Olga D. Lopina
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Maria A. Strelkova
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Iuliia D. Kuleshova
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Alexander A. Makarov
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Irina Yu. Petrushanko
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
2
|
Cordeiro BM, Leite Fontes CF, Meyer-Fernandes JR. Molecular Basis of Na, K-ATPase Regulation of Diseases: Hormone and FXYD2 Interactions. Int J Mol Sci 2024; 25:13398. [PMID: 39769162 PMCID: PMC11678576 DOI: 10.3390/ijms252413398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
The Na, K-ATPase generates an asymmetric ion gradient that supports multiple cellular functions, including the control of cellular volume, neuronal excitability, secondary ionic transport, and the movement of molecules like amino acids and glucose. The intracellular and extracellular levels of Na+ and K+ ions are the classical local regulators of the enzyme's activity. Additionally, the regulation of Na, K-ATPase is a complex process that occurs at multiple levels, encompassing its total cellular content, subcellular distribution, and intrinsic activity. In this context, the enzyme serves as a regulatory target for hormones, either through direct actions or via signaling cascades triggered by hormone receptors. Notably, FXYDs small transmembrane proteins regulators of Na, K-ATPase serve as intermediaries linking hormonal signaling to enzymatic regulation at various levels. Specifically, members of the FXYD family, particularly FXYD1 and FXYD2, are that undergo phosphorylation by kinases activated through hormone receptor signaling, which subsequently influences their modulation of Na, K-ATPase activity. This review describes the effects of FXYD2, cardiotonic steroid signaling, and hormones such as angiotensin II, dopamine, insulin, and catecholamines on the regulation of Na, K-ATPase. Furthermore, this review highlights the implications of Na, K-ATPase in diseases such as hypertension, renal hypomagnesemia, and cancer.
Collapse
Affiliation(s)
- Bárbara Martins Cordeiro
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
| | - Carlos Frederico Leite Fontes
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
| | - José Roberto Meyer-Fernandes
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
| |
Collapse
|
3
|
Trant J, Sanchez G, McDermott JP, Blanco G. Ouabain enhances renal cyst growth in a slowly progressive mouse model of autosomal dominant polycystic kidney disease. Am J Physiol Renal Physiol 2023; 325:F857-F869. [PMID: 37823195 PMCID: PMC10874652 DOI: 10.1152/ajprenal.00056.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 10/13/2023] Open
Abstract
Renal cyst progression in autosomal dominant polycystic kidney disease (ADPKD) is highly dependent on agents circulating in blood. We have previously shown, using different in vitro models, that one of these agents is the hormone ouabain. By binding to Na+-K+-ATPase (NKA), ouabain triggers a cascade of signal transduction events that enhance ADPKD cyst progression by stimulating cell proliferation, fluid secretion, and dedifferentiation of the renal tubular epithelial cells. Here, we determined the effects of ouabain in vivo. We show that daily administration of ouabain to Pkd1RC/RC ADPKD mice for 1-5 mo, at physiological levels, augmented kidney cyst area and number compared with saline-injected controls. Also, ouabain favored renal fibrosis; however, renal function was not significantly altered as determined by blood urea nitrogen levels. Ouabain did not have a sex preferential effect, with male and female mice being affected equally. By contrast, ouabain had no significant effect on wild-type mice. In addition, the actions of ouabain on Pkd1RC/RC mice were exacerbated when another mutation that increased the affinity of NKA for ouabain was introduced to the mice (Pkd1RC/RCNKAα1OS/OS mice). Altogether, this work highlights the role of ouabain as a procystogenic factor in the development of ADPKD in vivo, that the ouabain affinity site on NKA is critical for this effect, and that circulating ouabain is an epigenetic factor that worsens the ADPKD phenotype.NEW & NOTEWORTHY This work shows that the hormone ouabain enhances the progression of autosomal dominant polycystic kidney disease (ADPKD) in vivo. Ouabain augments the size and number of renal cysts, the kidney weight to body weight ratio, and kidney fibrosis in an ADPKD mouse model. The Na+-K+-ATPase affinity for ouabain plays a critical role in these effects. In addition, these outcomes are independent of the sex of the mice.
Collapse
Affiliation(s)
- Jordan Trant
- Department of Cell Biology and Physiology, University of Kansas Medical Center, The Kidney Institute, Kansas City, Kansas, United States
| | - Gladis Sanchez
- Department of Cell Biology and Physiology, University of Kansas Medical Center, The Kidney Institute, Kansas City, Kansas, United States
| | - Jeffrey P McDermott
- Department of Cell Biology and Physiology, University of Kansas Medical Center, The Kidney Institute, Kansas City, Kansas, United States
| | - Gustavo Blanco
- Department of Cell Biology and Physiology, University of Kansas Medical Center, The Kidney Institute, Kansas City, Kansas, United States
| |
Collapse
|
4
|
Current trends in natural products for the treatment and management of dementia: Computational to clinical studies. Neurosci Biobehav Rev 2023; 147:105106. [PMID: 36828163 DOI: 10.1016/j.neubiorev.2023.105106] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 02/24/2023]
Abstract
The number of preclinical and clinical studies evaluating natural products-based management of dementia has gradually increased, with an exponential rise in 2020 and 2021. Keeping this in mind, we examined current trends from 2016 to 2021 in order to assess the growth potential of natural products in the treatment of dementia. Publicly available literature was collected from various databases like PubMed and Google Scholar. Oxidative stress-related targets, NF-κB pathway, anti-tau aggregation, anti-AChE, and A-β aggregation were found to be common targets and pathways. A retrospective analysis of 33 antidementia natural compounds identified 125 sustainable resources distributed among 65 families, 39 orders, and 7 classes. We found that families such as Berberidaceae, Zingiberaceae, and Fabaceae, as well as orders such as Lamiales, Sapindales, and Myrtales, appear to be important and should be researched further for antidementia compounds. Moreover, some natural products, such as quercetin, curcumin, icariside II, berberine, and resveratrol, have a wide range of applications. Clinical studies and patents support the importance of dietary supplements and natural products, which we will also discuss. Finally, we conclude with the broad scope, future challenges, and opportunities for field researchers.
Collapse
|
5
|
Sharma R, Singh S, Tewari N, Dey P. A toxic shrub turned therapeutic: The dichotomy of Nerium oleander bioactivities. Toxicon 2023; 224:107047. [PMID: 36706925 DOI: 10.1016/j.toxicon.2023.107047] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/26/2023]
Abstract
Nerium oleander L. is a medicinal plant, used for the treatment of cancers and hyperglycemia across the world, especially in Indian sub-continent, Turkey, Morocco, and China. Although clinical studies supporting its pharmacological effects remain critically underexplored, accidental and intentional consumption of any part of the plant causes fatal toxicity in animals and humans. While the polyphenolic fraction of oleander leaves has been attributed to its pre-clinical pharmacological activities, the presence of diverse cardiac glycosides (especially oleandrin) causes apoptosis to cancer cells in vitro and results in clinical signs of oleander poisoning. Thus, the dual pharmacological and toxicological role of oleander is a perplexing dichotomy in phytotherapy. The current investigative review, therefore, intended to analyze the intrinsic and extrinsic factors that likely contribute to this conundrum. Especially by focusing on gut microbial diversity, abundance, and metabolic functions, oleander-associated pharmacological and toxicological studies have been critically analyzed to define the dual effects of oleander. Electronic databases were extensively screened for relevant research articles (including pre-clinical and clinical) related to oleander bioactivities and toxicity. Taxonomic preference was given to the plant N. oleander L. and synonymous plants as per 'The World Flora Online' database (WCSP record #135196). Discussion on yellow oleander (Cascabela thevetia (L.) Lippold) has intentionally been avoided since it is a different plant. The review indicates that the gut microbiota likely plays a key role in differentially modulating the pharmacological and toxicological effects of oleander. Other factors identified influencing the oleander bioactivities include dose and mode of treatment, cardiac glycoside pharmacokinetics, host-endogenous glycosides, plant material processing and phytochemical extraction methods, plant genotypic variations, environmental effects on the phytochemical quality and quantity, gene expression variations, host dietary patterns and co-morbidity, etc. The arguments proposed are also relevant to other medicinal plants containing toxic cardiac glycosides.
Collapse
Affiliation(s)
- Rajat Sharma
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India.
| | - Swati Singh
- Department of Zoology, University of North Bengal, Siliguri, West Bengal, India.
| | - Nisha Tewari
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India.
| | - Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India.
| |
Collapse
|
6
|
Takada Y, Kaneko K, Kawakami Y. Interaction of Odoroside A, A Known Natural Cardiac Glycoside, with Na +/K +-ATPase. J Membr Biol 2023; 256:229-241. [PMID: 36840763 DOI: 10.1007/s00232-023-00281-1] [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: 09/21/2022] [Accepted: 02/01/2023] [Indexed: 02/26/2023]
Abstract
The nature of odoroside A, a cardiac glycoside (CG) extracted from Nerium oleander, as well as its chemical structure is quite similar to a well-known CG, ouabain possessing a steroid skeleton, a five-membered unsaturated lactone ring, and a sugar moiety as a common structure. Like ouabain, odoroside A inhibits the activity of Na+/K+-ATPase (NKA) and shows significant anticancer activity, however its inhibitory mechanism remains unknown. CGs show various physiological activities, including cardiotonic and anticancer activities, through the inhibition of NKA by direct interaction. Additionally, X-ray crystallographic analysis revealed the inhibitory mechanism of ouabain and digoxin in relation to NKA. By using different molecular modeling techniques, docking simulation of odoroside A and NKA was conducted based on the results of these X-ray crystallographic analyses. Furthermore, a comparison of the results with the binding characteristics of three known CGs (ouabain, digoxin, and digitoxin) was also conducted. Odoroside A fitted into the CG binding pocket on the α-subunit of NKA revealed by X-ray crystallography. It had key interactions with Thr797 and Phe783. Also, three known CGs showed similar interactions with Thr797 and Phe783. Interaction modes of odoroside A were quite similar to those of ouabain, digoxin, and digitoxin. Docking simulations indicated that the sugar moiety enhanced the interaction between NKA and CGs, but did not show enhanced NKA inhibitory activity because the sugar moiety was placed outside the entrance of active site. Thus, these results suggest that the inhibitory mechanism of odoroside A to NKA is the same as the known CGs.
Collapse
Affiliation(s)
- Yohei Takada
- Corporate Planning Department, Otsuka Holdings Co., Ltd, Shinagawa Grand Central Tower 2-16-4 Konan, Minato-Ku, Tokyo, 108-8241, Japan.
| | - Kazuhiro Kaneko
- Headquarters of Clinical Development, Otsuka Pharmaceutical Co., Ltd, Shinagawa Grand Central Tower 2-16-4 Konan, Minato-Ku, Tokyo, 108-8241, Japan
| | | |
Collapse
|
7
|
Chasalow F. An Introduction to Spiral Steroids. Int J Mol Sci 2022; 23:ijms23179523. [PMID: 36076935 PMCID: PMC9455587 DOI: 10.3390/ijms23179523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/13/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
In addition to classical steroids, which have cholesterol as a precursor, there are steroids with 7-dehydrocholesterol as a precursor. This review describes the identification of these steroids, their biosynthesis, and some aspects of their function. There are three classes of these compounds, distinguished by the number of their carbon atoms, 23, 24, and 25. Each class has a spiral steroid and is a phosphodiester. Up until these investigations, no spiral steroids or steroid phosphodiesters were known. There are at least 13 compounds, of which six have been purified to near homogeneity; each one has been characterized by its mass and proposed composition, and they function by regulating the NaK-ATPase. Based on the tissues in which they have been detected, each class of compound seems to regulate a different isoform of the NaK-ATPase. This is an important site of endocrine regulation.
Collapse
Affiliation(s)
- Fred Chasalow
- IOMA LLC, Belmont, CA 94002-3321, USA; ; Tel.: +1-650-576-1800
- Department of Laboratory Medicine, VAMC, San Francisco, CA 94121-1545, USA
| |
Collapse
|
8
|
da Silva Ferreira R, Fernandes PBU, da Cruz JPO, Silva FLA, Lempek MR, Canta GN, Veado JCC, Mantovani MM, Botelho AFM, Melo MM. Comparative Therapeutic Potential of Cardioactive Glycosides in Doxorubicin Model of Heart Failure. Cardiovasc Toxicol 2022; 22:78-87. [PMID: 34655414 DOI: 10.1007/s12012-021-09702-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/01/2021] [Indexed: 11/28/2022]
Abstract
In the present study, we investigated the cardioactive glycosides oleandrin and ouabain, and compared them to digoxin in a model of cardiotoxicity induced by doxorubicin. Adult rats were distributed into four experimental groups. Each group was challenged with a single intraperitoneal application of doxorubicin at a dose of 12 mg/kg. Then, they were treated with saline solution and the glycosides oleandrin, ouabain, and digoxin at a dose of 50 µg/kg, for 7 days. They underwent echocardiography, electrocardiography, hematologic, biochemical tests, and microscopic evaluation of the heart. All animals presented congestive heart failure, which was verified by a reduction in the ejection fraction. Oleandrin and digoxin were able to significantly reduce (p < 0.05) the eccentric remodeling caused by doxorubicin. Oleandrin and digoxin were significantly lower (p < 0.05) than the control group in maintaining systolic volume and left ventricular volume in diastole. Other parameters evaluated did not show significant statistical differences. All animals showed an increase in erythrocyte count, and an increase in the duration of the QRS complex on the ECG and myocardial necrosis at the histopathological analysis. It is concluded that the glycosides oleandrin, ouabain, and digoxin in the used dosage do not present therapeutic potential for the treatment of congestive heart failure caused by doxorubicin.
Collapse
Affiliation(s)
| | | | | | | | - Marthin Raboch Lempek
- Escola de Veterinária da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Gioavanni Naves Canta
- Instituto de Ciência Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | - Marília Martins Melo
- Escola de Veterinária da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
9
|
Chen L, Zhang M, Wang X, Liu Y, Bian J, Yan D, Yin W. Cardiac steroid ouabain transcriptionally increases human leukocyte antigen DR expression on monocytes. Steroids 2021; 175:108915. [PMID: 34508735 DOI: 10.1016/j.steroids.2021.108915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/31/2021] [Accepted: 08/30/2021] [Indexed: 01/18/2023]
Abstract
Sepsis is a life-threatening disease characterized by acute multiple organ dysfunction and immunosuppression that is also called as immunoparalysis. Increasing evidence suggests that immunoparalysis largely contributes to the high mortality of sepsis, but the effective remedies are lacking. As an important antigen presentation molecule, human leukocyte antigen DR (HLA-DR) is remarkably down-regulated in sepsis-induced immunoparalysis, therefore, re-stimulation of HLA-DR expression is expected to be useful in reversing immunoparalysis. We previously described that ouabain, as a Na+, K+-ATPase ligand, is able to counteract immunoparalysis by regulating TH1 cytokines expression. Here, we expanded the finding that ouabain not only prevents LPS-induced down-regulation of HLA-DR on monocytes, but also transcriptionally activates HLA-DR α/β expression mediated by CIITA4, IRF1, c-Src, and Stat1 phosphorylation. Since ouabain can improve sepsis-induced immunoparalysis by multiple mechanisms, we propose that ouabain may be a promising agent in septic therapy that deserves further investigation.
Collapse
Affiliation(s)
- Lili Chen
- State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
| | - Manli Zhang
- State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
| | - Xiya Wang
- State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
| | - Yongjian Liu
- State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
| | - Jinjun Bian
- Department of Anesthesiology and Critical Care, Changhai Hospital, Naval Medical University, Shanghai, China.
| | - Dong Yan
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, China.
| | - Wu Yin
- State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China.
| |
Collapse
|
10
|
Dey P. The pharmaco-toxicological conundrum of oleander: Potential role of gut microbiome. Biomed Pharmacother 2020; 129:110422. [PMID: 32563990 DOI: 10.1016/j.biopha.2020.110422] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/08/2020] [Accepted: 06/14/2020] [Indexed: 02/08/2023] Open
Abstract
Nerium oleander L., commonly known as oleander, is a toxic shrub and also a medicinal plant. All parts of oleander are rich in cardiac glycosides that inhibits Na+/K+-ATPase and induce inotropic effect on the cardiomyocytes. Several pre-clinical and clinical reports indicate acute toxicity due to intentional, accidental and suicidal oleander consumption. Contrarily, oleander is used for the treatment of diverse ailments in traditional medicinal practices around the globe and several evidence-based pre-clinical studies indicated metabolic and immunological health benefits of polyphenol-rich oleander extracts. Thus, the current review aims to address this pharmaco-toxicological conundrum of oleander by addressing the possible role of gut microflora in the differential oleander toxicity. Additionally, a comprehensive account of ethnopharmacological usage, metabolic and immunological health benefits has been documented that supplement the conflicting arguments of pharmaco-toxicological properties of oleander. Finally, by addressing the gap of knowledge of ethnomedicinal, pharmacological and toxicological reports of oleander, the current review is expected to pave the way to address the differential pharmaco-toxicological effects of oleander.
Collapse
Affiliation(s)
- Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India.
| |
Collapse
|
11
|
Elucidating Potential Profibrotic Mechanisms of Emerging Biomarkers for Early Prognosis of Hepatic Fibrosis. Int J Mol Sci 2020; 21:ijms21134737. [PMID: 32635162 PMCID: PMC7369895 DOI: 10.3390/ijms21134737] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatic fibrosis has been associated with a series of pathophysiological processes causing excessive accumulation of extracellular matrix proteins. Several cellular processes and molecular mechanisms have been implicated in the diseased liver that augments fibrogenesis, fibrogenic cytokines and associated liver complications. Liver biopsy remains an essential diagnostic tool for histological evaluation of hepatic fibrosis to establish a prognosis. In addition to being invasive, this methodology presents with several limitations including poor cost-effectiveness, prolonged hospitalizations, and risks of peritoneal bleeding, while the clinical use of this method does not reveal underlying pathogenic mechanisms. Several alternate noninvasive diagnostic strategies have been developed, to determine the extent of hepatic fibrosis, including the use of direct and indirect biomarkers. Immediate diagnosis of hepatic fibrosis by noninvasive means would be more palatable than a biopsy and could assist clinicians in taking early interventions timely, avoiding fatal complications, and improving prognosis. Therefore, we sought to review some common biomarkers of liver fibrosis along with some emerging candidates, including the oxidative stress-mediated biomarkers, epigenetic and genetic markers, exosomes, and miRNAs that needs further evaluation and would have better sensitivity and specificity. We also aim to elucidate the potential role of cardiotonic steroids (CTS) and evaluate the pro-inflammatory and profibrotic effects of CTS in exacerbating hepatic fibrosis. By understanding the underlying pathogenic processes, the efficacy of these biomarkers could allow for early diagnosis and treatment of hepatic fibrosis in chronic liver diseases, once validated.
Collapse
|
12
|
Chang YM, Shih YL, Chen CP, Liu KL, Lee MH, Lee MZ, Hou HT, Huang HC, Lu HF, Peng SF, Chen KW, Yeh MY, Chung JG. Ouabain induces apoptotic cell death in human prostate DU 145 cancer cells through DNA damage and TRAIL pathways. ENVIRONMENTAL TOXICOLOGY 2019; 34:1329-1339. [PMID: 31436044 DOI: 10.1002/tox.22834] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/01/2019] [Accepted: 08/02/2019] [Indexed: 06/10/2023]
Abstract
Ouabain, a cardiotonic steroid and specific Na+ /K+ -ATPase inhibitor, has a potential to induce cancer cell apoptosis but the mechanisms of apoptosis induced by ouabain are not fully understand. The aim of this study was to investigate the cytotoxic effects of ouabain on human prostate cancer DU 145 cells in vitro. Cell morphological changes were examined by phase contrast microscopy. Cell viability, cell cycle distribution, cell apoptosis, DNA damage, the production of ROS and Ca2+ , and mitochondrial membrane potential (ΔΨm ) were measured by flow cytometry assay. Results indicated that ouabain induced cell morphological changes, decreased total cell viability, induced G0/G1 phase arrest, DNA damage, and cell apoptosis, increased ROS and Ca2+ production, but decreased the levels of ΔΨm in DU 145 cells. Ouabain also increased the activities of caspase-3, -8, and -9. Western blotting was used for measuring the alterations of apoptosis-associated protein expressions in DU 145 cells and results indicated that ouabain increased the expression of DNA damage associated proteins (pATMSer1981 , p-H2A.XSer139 , and p-p53Ser15 ) and ER-stress-associated proteins (Grp78, ATF6β, p-PERKThr981 , PERK, eIF2A, GADD153, CaMKIIβ, and caspase-4) in time-dependently. Furthermore, ouabain increased apoptosis-associated proteins (DR4, DR5, Fas, Fas Ligand, and FADD), TRAIL pathway, which related to extrinsic pathway, promoted the pro-apoptotic protein Bax, increased apoptotic-associated proteins, such as cytochrome c, AIF, Endo G, caspase-3, -8, and -9, but reduced anti-apoptotic protein Bcl-2 and Bcl-x in DU 145 cells. In conclusion, we may suggest that ouabain decreased cell viability and induced apoptotic cell death may via caspase-dependent and mitochondria-dependent pathways in human prostate cancer DU 145 cells.
Collapse
Affiliation(s)
- Yi-Ming Chang
- Department of Pathology, Tri-service General Hospital and Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Yung-Luen Shih
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei, Taiwan
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei, Taiwan
| | - Chao-Ping Chen
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Ko-Lin Liu
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Mei-Hui Lee
- Department of Genetic Counseling Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Ming-Zhe Lee
- Department of Clinical Pathology, Cheng-Hsin General Hospital, Taipei, Taiwan
| | - Hsin-Tu Hou
- Department of Clinical Pathology, Cheng-Hsin General Hospital, Taipei, Taiwan
| | - Hsieh-Chou Huang
- Department of Anesthesiology and Pain Medicine, Cheng-Hsin General Hospital, Taipei, Taiwan
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| | - Hsu-Feng Lu
- Department of Clinical Pathology, Cheng-Hsin General Hospital, Taipei, Taiwan
- Department of Restaurant, Hotel and Institutional Management, Fu-Jen Catholic University, New Taipei, Taiwan
| | - Shu-Fen Peng
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Kuo-Wei Chen
- Division of Hematology and Oncology, Cheng-Hsin General Hospital, Taipei, Taiwan
| | - Ming-Yang Yeh
- Department of Education and Research, Cheng-Hsin General Hospital, Taipei, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
13
|
Lu J, Wang HW, Ahmad M, Keshtkar-Jahromi M, Blaustein MP, Hamlyn JM, Leenen FHH. Central and peripheral slow-pressor mechanisms contributing to Angiotensin II-salt hypertension in rats. Cardiovasc Res 2019; 114:233-246. [PMID: 29126194 DOI: 10.1093/cvr/cvx214] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/07/2017] [Indexed: 12/17/2022] Open
Abstract
Aims High salt intake markedly enhances hypertension induced by angiotensin II (Ang II). We explored central and peripheral slow-pressor mechanisms which may be activated by Ang II and salt. Methods and results In protocol I, Wistar rats were infused subcutaneously with low-dose Ang II (150 ng/kg/min) and fed regular (0.4%) or high salt (2%) diet for 14 days. In protocol II, Ang II-high salt was combined with intracerebroventricular infusion of mineralocorticoid receptor (MR) blockers (eplerenone, spironolactone), epithelial sodium channel (ENaC) blocker (benzamil), angiotensin II type 1 receptor (AT1R) blocker (losartan) or vehicles. Ang II alone raised mean arterial pressure (MAP) ∼10 mmHg, but Ang II-high salt increased MAP ∼50 mmHg. Ang II-high salt elevated plasma corticosterone, aldosterone and endogenous ouabain but not Ang II alone. Both Ang II alone and Ang II-high salt increased mRNA and protein expression of CYP11B2 (aldosterone synthase gene) in the adrenal cortex but not of CYP11B1 (11-β-hydroxylase gene). In the aorta, Ang II-high salt increased sodium-calcium exchanger-1 (NCX1) protein. The Ang II-high salt induced increase in MAP was largely prevented by central infusion of MR blockers, benzamil or losartan. Central blockades significantly lowered plasma aldosterone and endogenous ouabain and markedly decreased Ang II-high salt induced CYP11B2 mRNA expression in the adrenal cortex and NCX1 protein in the aorta. Conclusion These results suggest that in Ang II-high salt hypertension, MR-ENaC-AT1R signalling in the brain increases circulating aldosterone and endogenous ouabain, and arterial NCX1. These factors can amplify blood pressure responses to centrally-induced sympatho-excitation and thereby contribute to severe hypertension.
Collapse
Affiliation(s)
- Jiao Lu
- Brain and Heart Research Group, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y 4W7, Canada
| | - Hong-Wei Wang
- Brain and Heart Research Group, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y 4W7, Canada
| | - Monir Ahmad
- Brain and Heart Research Group, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y 4W7, Canada
| | - Marzieh Keshtkar-Jahromi
- Brain and Heart Research Group, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y 4W7, Canada.,Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore S, Baltimore, MD 21201, USA
| | - Mordecai P Blaustein
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore S, Baltimore, MD 21201, USA.,Department of Medicine, University of Maryland School of Medicine, 655 West Baltimore S, Baltimore, MD 21201, USA
| | - John M Hamlyn
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore S, Baltimore, MD 21201, USA
| | - Frans H H Leenen
- Brain and Heart Research Group, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y 4W7, Canada
| |
Collapse
|
14
|
Vilchis-Nestor CA, Roldán ML, Leonardi A, Navea JG, Padilla-Benavides T, Shoshani L. Ouabain Enhances Cell-Cell Adhesion Mediated by β 1 Subunits of the Na +,K +-ATPase in CHO Fibroblasts. Int J Mol Sci 2019; 20:E2111. [PMID: 31035668 PMCID: PMC6539428 DOI: 10.3390/ijms20092111] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 12/19/2022] Open
Abstract
Adhesion is a crucial characteristic of epithelial cells to form barriers to pathogens and toxic substances from the environment. Epithelial cells attach to each other using intercellular junctions on the lateral membrane, including tight and adherent junctions, as well as the Na+,K+-ATPase. Our group has shown that non-adherent chinese hamster ovary (CHO) cells transfected with the canine β1 subunit become adhesive, and those homotypic interactions amongst β1 subunits of the Na+,K+-ATPase occur between neighboring epithelial cells. Ouabain, a cardiotonic steroid, binds to the α subunit of the Na+,K+-ATPase, inhibits the pump activity and induces the detachment of epithelial cells when used at concentrations above 300 nM. At nanomolar non-inhibiting concentrations, ouabain affects the adhesive properties of epithelial cells by inducing the expression of cell adhesion molecules through the activation of signaling pathways associated with the α subunit. In this study, we investigated whether the adhesion between β1 subunits was also affected by ouabain. We used CHO fibroblasts stably expressing the β1 subunit of the Na+,K+-ATPase (CHO β1), and studied the effect of ouabain on cell adhesion. Aggregation assays showed that ouabain increased the adhesion between CHO β1 cells. Immunofluorescence and biotinylation assays showed that ouabain (50 nM) increases the expression of the β1 subunit of the Na+,K+-ATPase at the cell membrane. We also examined the effect of ouabain on the activation of signaling pathways in CHO β1 cells, and their subsequent effect on cell adhesion. We found that cSrc is activated by ouabain and, therefore, that it likely regulates the adhesive properties of CHO β1 cells. Collectively, our findings suggest that the β1 subunit adhesion is modulated by the expression levels of the Na+,K+-ATPase at the plasma membrane, which is regulated by ouabain.
Collapse
Affiliation(s)
- Claudia Andrea Vilchis-Nestor
- Department of Physiology Biophysics and Neurosciences, Center for Research and Advanced Studies, Cinvestav-Ipn, CDMX 07360, Mexico.
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - María Luisa Roldán
- Department of Physiology Biophysics and Neurosciences, Center for Research and Advanced Studies, Cinvestav-Ipn, CDMX 07360, Mexico.
| | - Angelina Leonardi
- Department of Chemistry, Skidmore College, 815 North Broadway, Saratoga Springs, NY 12866, USA.
| | - Juan G Navea
- Department of Chemistry, Skidmore College, 815 North Broadway, Saratoga Springs, NY 12866, USA.
| | - Teresita Padilla-Benavides
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Liora Shoshani
- Department of Physiology Biophysics and Neurosciences, Center for Research and Advanced Studies, Cinvestav-Ipn, CDMX 07360, Mexico.
| |
Collapse
|
15
|
Simonini M, Casanova P, Citterio L, Messaggio E, Lanzani C, Manunta P. Reply: "Comment on: Endogenous Ouabain and Related Genes in the Translation from Hypertension to Renal Diseases". Int J Mol Sci 2019; 20:ijms20030542. [PMID: 30696018 PMCID: PMC6387140 DOI: 10.3390/ijms20030542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 01/17/2019] [Indexed: 12/31/2022] Open
Affiliation(s)
- Marco Simonini
- Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Università Vita Salute San Raffaele, 20132 Milan, Italy.
| | - Paola Casanova
- Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Università Vita Salute San Raffaele, 20132 Milan, Italy.
| | - Lorena Citterio
- Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Università Vita Salute San Raffaele, 20132 Milan, Italy.
| | - Elisabetta Messaggio
- Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Università Vita Salute San Raffaele, 20132 Milan, Italy.
| | - Chiara Lanzani
- Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Università Vita Salute San Raffaele, 20132 Milan, Italy.
| | - Paolo Manunta
- Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Università Vita Salute San Raffaele, 20132 Milan, Italy.
| |
Collapse
|
16
|
Song HL, Demirev AV, Kim NY, Kim DH, Yoon SY. Ouabain activates transcription factor EB and exerts neuroprotection in models of Alzheimer's disease. Mol Cell Neurosci 2018; 95:13-24. [PMID: 30594669 DOI: 10.1016/j.mcn.2018.12.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/20/2018] [Accepted: 12/26/2018] [Indexed: 11/24/2022] Open
Abstract
The number of neurofibrillary tangles containing abnormal hyperphosphorylated tau protein correlates with the degree of dementia in Alzheimer's disease (AD). In addition, autophagosome accumulation and disturbance of autophagy, the process by which toxic aggregate proteins are degraded in the cytosol, are also found in AD models. These indicate that regulation of the autophagy-lysosome system may be a potential therapeutic target for AD. Activation of transcription factor EB (TFEB), a master regulator of autophagy-lysosome system gene transcription, reduces the amount of tau in APP mice. Here, to identify potential therapeutic compounds for AD, we performed two types of screening to determine pharmacologically active compounds that increase 1) neuronal viability in okadaic acid-induced tau hyperphosphorylation-related neurodegeneration models and 2) nuclear localization of TFEB in high-contents screening. Ouabain, a cardiac glycoside, was discovered as a common hit compound in both screenings. It also exhibited a significant protective effect in tau transgenic fly and mouse models in vivo. This work demonstrates that ouabain enhances activation of TFEB through inhibition of the mTOR pathway and induces downstream autophagy-lysosomal gene expression and cellular restorative properties. Therefore, therapeutic approaches using ouabain reduce the accumulation of abnormal toxic tau in vitro and in vivo.
Collapse
Affiliation(s)
- Ha-Lim Song
- Department of Brain Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Bio-Medical Institute of Technology (BMIT), University of Ulsan College of Medicine, Seoul, Republic of Korea; Institute for Innovation in Neurodegenerative Diseases, ADEL, Inc., Seoul, Republic of Korea
| | - Atanas Vladimirov Demirev
- Department of Brain Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Bio-Medical Institute of Technology (BMIT), University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Na-Young Kim
- Department of Brain Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Bio-Medical Institute of Technology (BMIT), University of Ulsan College of Medicine, Seoul, Republic of Korea; Institute for Innovation in Neurodegenerative Diseases, ADEL, Inc., Seoul, Republic of Korea
| | - Dong-Hou Kim
- Department of Brain Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Bio-Medical Institute of Technology (BMIT), University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Seung-Yong Yoon
- Department of Brain Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Bio-Medical Institute of Technology (BMIT), University of Ulsan College of Medicine, Seoul, Republic of Korea; Institute for Innovation in Neurodegenerative Diseases, ADEL, Inc., Seoul, Republic of Korea.
| |
Collapse
|
17
|
Role of endogenous digitalis-like factors in the clinical manifestations of severe preeclampsia: a sytematic review. Clin Sci (Lond) 2018; 132:1215-1242. [PMID: 29930141 DOI: 10.1042/cs20171499] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 04/26/2018] [Accepted: 05/08/2018] [Indexed: 12/18/2022]
Abstract
Endogenous digitalis-like factor(s), originally proposed as a vasoconstrictor natriuretic hormone, was discovered in fetal and neonatal blood accidentally because it cross-reacts with antidigoxin antibodies (ADAs). Early studies using immunoassays with ADA identified the digoxin-like immuno-reactive factor(s) (EDLF) in maternal blood as well, and suggested it originated in the feto-placental unit. Mammalian digoxin-like factors have recently been identified as at least two classes of steroid compounds, plant derived ouabain (O), and several toad derived bufodienolides, most prominent being marinobufagenin (MBG). A synthetic pathway for MBG has been identified in mammalian placental tissue. Elevated maternal and fetal EDLF, O and MBG have been demonstrated in preeclampsia (PE), and inhibition of red cell membrane sodium, potassium ATPase (Na, K ATPase (NKA)) by EDLF is reversed by ADA fragments (ADA-FAB). Accordingly, maternal administration of a commercial ADA-antibody fragment (FAB) was tested in several anecdotal cases of PE, and two, small randomized, prospective, double-blind clinical trials. In the first randomized trial, ADA-FAB was administered post-partum, in the second antepartum. In the post-partum trial, ADA-FAB reduced use of antihypertensive drugs. In the second trial, there was no effect of ADA-FAB on blood pressure, but the fall in maternal creatinine clearance (CrCl) was prevented. In a secondary analysis using the pre-treatment maternal level of circulating Na, K ATPase (NKA) inhibitory activity (NKAI), ADA-FAB reduced the incidence of pulmonary edema and, unexpectedly, that of severe neonatal intraventricular hemorrhage (IVH). The fall in CrCl in patients given placebo was proportional to the circulating level of NKAI. The implications of these findings on the pathophysiology of the clinical manifestations PE are discussed, and a new model of the respective roles of placenta derived anti-angiogenic (AAG) factors (AAGFs) and EDLF is proposed.
Collapse
|
18
|
Abstract
Studies in the early 1990s suggested that a hormone identical to ouabain or an isomer of ouabain is secreted by the adrenal glands into the circulation and plays a role in the regulation of arterial pressure and cardiac and renal function. This hormone, known as endogenous ouabain (EO), was claimed to contribute to the pathophysiology of a number of disorders including heart failure, renal failure, pregnancy-induced, and essential hypertension. However, some research groups have been unable to confirm the presence of EO in the human circulation and the issue remains in dispute. In that the implications are of considerable importance to clinicians who, like the authors, lack biochemical expertise, it would be useful if the dispute could be addressed by disinterested scientists with long-standing and acknowledged expertise in analytical chemistry who could opine as to whether the evidence is, or is not, sufficient to state categorically that EO does (or does not) exist in the circulation in man. This brief review does not present new data but, rather, recommends that adjudication is needed regarding this important issue. © 2018 BioFactors, 44(3):219-221, 2018.
Collapse
Affiliation(s)
- Risto J Kaaja
- Institute of Clinical Medicine, Internal Medicine, University of Turku and Turku University Hospital, Finland
| | - M Gary Nicholls
- Christchurch Hospital, University of Otago-Christchurch, Christchurch, New Zealand
| |
Collapse
|
19
|
Bhattarai B, Nagorny P. Enantioselective Total Synthesis of Cannogenol-3-O-α-l-rhamnoside via Sequential Cu(II)-Catalyzed Michael Addition/Intramolecular Aldol Cyclization Reactions. Org Lett 2017; 20:154-157. [PMID: 29244520 DOI: 10.1021/acs.orglett.7b03513] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A concise and scalable enantioselective total synthesis of the natural cardenolides cannogenol and cannogenol-3-O-α-l-rhamnoside has been achieved in 18 linear steps. The synthesis features a Cu(II)-catalyzed enantioselective and diastereoselective Michael reaction/tandem aldol cyclization and a one-pot reduction/transposition, which resulted in a rapid (6 linear steps) assembly of a functionalized intermediate containing C19 oxygenation that could be elaborated to cardenolide cannogenol. In addition, a strategy for achieving regio- and stereoselective glycosylation at the C3 position of synthetic cannogenol was developed and applied to the preparation of cannogenol-3-O-α-l-rhamnoside.
Collapse
Affiliation(s)
- Bijay Bhattarai
- Chemistry Department, University of Michigan , 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| | - Pavel Nagorny
- Chemistry Department, University of Michigan , 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
20
|
Cavalcante-Silva LHA, Lima ÉDA, Carvalho DCM, de Sales-Neto JM, Alves AKDA, Galvão JGFM, da Silva JSDF, Rodrigues-Mascarenhas S. Much More than a Cardiotonic Steroid: Modulation of Inflammation by Ouabain. Front Physiol 2017; 8:895. [PMID: 29176951 PMCID: PMC5686084 DOI: 10.3389/fphys.2017.00895] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 10/24/2017] [Indexed: 12/22/2022] Open
Abstract
Since the discovery of ouabain as a cardiotonic steroid hormone present in higher mammals, research about it has progressed rapidly and several of its physiological and pharmacological effects have been described. Ouabain can behave as a stress hormone and adrenal cortex is its main source. Direct effects of ouabain are originated due to the binding to its receptor, the Na+/K+-ATPase, on target cells. This interaction can promote Na+ transport blockade or even activation of signaling transduction pathways (e.g., EGFR/Src-Ras-ERK pathway activation), independent of ion transport. Besides the well-known effect of ouabain on the cardiovascular system and blood pressure control, compelling evidence indicates that ouabain regulates a number of immune functions. Inflammation is a tightly coordinated immunological function that is also affected by ouabain. Indeed, this hormone can modulate many inflammatory events such as cell migration, vascular permeability, and cytokine production. Moreover, ouabain also interferes on neuroinflammation. However, it is not clear how ouabain controls these events. In this brief review, we summarize the updates of ouabain effect on several aspects of peripheral and central inflammation, bringing new insights into ouabain functions on the immune system.
Collapse
Affiliation(s)
- Luiz H A Cavalcante-Silva
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Laboratório de Imunobiotecnologia, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Éssia de Almeida Lima
- Programa de Pós-Graduação em Biotecnologia, Laboratório de Imunobiotecnologia, Centro de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Deyse C M Carvalho
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Laboratório de Imunobiotecnologia, Centro de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - José M de Sales-Neto
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Laboratório de Imunobiotecnologia, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Anne K de Abreu Alves
- Programa de Pós-Graduação em Biotecnologia, Laboratório de Imunobiotecnologia, Centro de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - José G F M Galvão
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Laboratório de Imunobiotecnologia, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Juliane S de França da Silva
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Laboratório de Imunobiotecnologia, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Sandra Rodrigues-Mascarenhas
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Laboratório de Imunobiotecnologia, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil.,Programa de Pós-Graduação em Biotecnologia, Laboratório de Imunobiotecnologia, Centro de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, Brazil.,Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Laboratório de Imunobiotecnologia, Centro de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, Brazil
| |
Collapse
|
21
|
Endogenous ouabain and aldosterone are coelevated in the circulation of patients with essential hypertension. J Hypertens 2017; 34:2074-80. [PMID: 27457665 DOI: 10.1097/hjh.0000000000001042] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE In the setting of normal sodium (Na) intake, many patients with hypertension have inappropriately elevated plasma aldosterone (Aldo) levels and may be at increased risk for tissue damage. Moreover, other adrenocortical steroids, including endogenous ouabain can stimulate tissue damage. As endogenous ouabain is often elevated in chronically Na-loaded states, is a vasoconstrictor, raises blood pressure (BP), and also promotes tissue fibrosis, we investigated the extent to which plasma Aldo and endogenous ouabain were coelevated among naïve hypertensive patients (NHP). We also investigated the impact of an acute salt load on these steroids, BP, and renal function. METHODS NHP (590) were grouped in tertiles based on their baseline plasma Aldo (mean ± SEM first 7.59 ± 0.18, versus third 24.15 ± 0.31 ng/dl). Baseline plasma renin activity (2.4 ± 0.1 versus 1.2 ± 0.1 ng/ml per h, P < 0.001), endogenous ouabain (268 ± 14.9 pmol/l versus 239.0 ± 13.6 pmol, P < 0.01) and DBP (91.9 ± 0.76 versus 89.6 ± 0.71 mmHg, P = 0.017) were higher in NHP in the third versus the first Aldo tertile, respectively. RESULTS Acute Na loading showed that the BP of the third Aldo tertile NHP was especially salt-sensitive (slope of pressure-natriuresis relationship 0.015 ± 0.002 versus 0.003 ± 0.001 μEq/mmHg per min, P = 0.00024 after adjustment for sex, BMI, and age). Regression analyses showed that plasma Aldo and endogenous ouabain were linearly related (β = 0.181, P = 0.0003). CONCLUSION Among patients with essential hypertension, circulating endogenous ouabain and Aldo are typically coelevated and their BP is salt-sensitive. In conditions where Aldo is inappropriately elevated, both Aldo and endogenous ouabain may contribute to adverse cardiovascular and renal outcomes.
Collapse
|
22
|
Mohammadi S, French SS, Neuman-Lee LA, Durham SL, Kojima Y, Mori A, Brodie ED, Savitzky AH. Corticosteroid responses of snakes to toxins from toads (bufadienolides) and plants (cardenolides) reflect differences in dietary specializations. Gen Comp Endocrinol 2017; 247:16-25. [PMID: 28347742 DOI: 10.1016/j.ygcen.2017.03.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/07/2017] [Accepted: 03/22/2017] [Indexed: 01/24/2023]
Abstract
Toads are chemically defended by cardiotonic steroids known as bufadienolides. Resistance to the acute effects of bufadienolides in snakes that prey on toads is conferred by target-site insensitivity of the toxin's target enzyme, the Na+/K+-ATPase. Previous studies have focused largely on the molecular mechanisms of resistance but have not investigated the physiological mechanisms or consequences of exposure to the toxins. Adrenal enlargement in snakes often is associated with specialization on a diet of toads. These endocrine glands are partly composed of interrenal tissue, which produces the corticosteroids corticosterone and aldosterone. Corticosterone is the main hormone released in response to stress in reptiles, and aldosterone plays an important role in maintaining ion balance through upregulation of Na+/K+-ATPase. We tested the endocrine response of select species of snakes to acute cardiotonic steroid exposure by measuring circulating aldosterone and corticosterone concentrations. We found that Rhabdophis tigrinus, which specializes on a diet of toads, responds with lower corticosterone and higher aldosterone compared to other species that exhibit target-site resistance to the toxins but do not specialize on toads. We also found differences between sexes in R. tigrinus, with males generally responding with higher corticosterone and aldosterone than females. This study provides evidence of physiological adaptations, beyond target-site resistance, associated with tolerance of bufadienolides in a specialized toad-eating snake.
Collapse
Affiliation(s)
- Shabnam Mohammadi
- Department of Biology, Utah State University, 5305 Old Main Hill, Logan, UT 84322-5305, United States; Ecology Center, Utah State University, 5205 Old Main Hill, Logan, UT 84322-5205, United States.
| | - Susannah S French
- Department of Biology, Utah State University, 5305 Old Main Hill, Logan, UT 84322-5305, United States; Ecology Center, Utah State University, 5205 Old Main Hill, Logan, UT 84322-5205, United States
| | - Lorin A Neuman-Lee
- Department of Biology, Utah State University, 5305 Old Main Hill, Logan, UT 84322-5305, United States
| | - Susan L Durham
- Ecology Center, Utah State University, 5205 Old Main Hill, Logan, UT 84322-5205, United States
| | - Yosuke Kojima
- Department of Zoology, Graduate School of Science, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Akira Mori
- Department of Zoology, Graduate School of Science, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Edmund D Brodie
- Department of Biology, Utah State University, 5305 Old Main Hill, Logan, UT 84322-5305, United States; Ecology Center, Utah State University, 5205 Old Main Hill, Logan, UT 84322-5205, United States
| | - Alan H Savitzky
- Department of Biology, Utah State University, 5305 Old Main Hill, Logan, UT 84322-5305, United States; Ecology Center, Utah State University, 5205 Old Main Hill, Logan, UT 84322-5205, United States
| |
Collapse
|
23
|
Coccini T, Caloni F, De Simone U. Human neuronal cell based assay: A new in vitro model for toxicity evaluation of ciguatoxin. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 52:200-213. [PMID: 28437641 DOI: 10.1016/j.etap.2017.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 03/31/2017] [Accepted: 04/02/2017] [Indexed: 06/07/2023]
Abstract
Ciguatoxins (CTXs) are emerging marine neurotoxins representing the main cause of ciguatera fish poisoning, an intoxication syndrome which configures a health emergency and constitutes an evolving issue constantly changing due to new vectors and derivatives of CTXs, as well as their presence in new non-endemic areas. The study applied the neuroblastoma cell model of human origin (SH-SY5Y) to evaluate species-specific mechanistic information on CTX toxicity. Metabolic functionality, cell morphology, cytosolic Ca2+i responses, neuronal cell growth and proliferation were assessed after short- (4-24h) and long-term exposure (10days) to P-CTX-3C. In SH-SY5Y, P-CTX-3C displayed a powerful cytotoxicity requiring the presence of both Veratridine and Ouabain. SH-SY5Y were very sensitive to Ouabain: 10 and 0.25nM appeared the optimal concentrations, for short- and long-term toxicity studies, respectively, to be used in co-incubation with Veratridine (25μM), simulating the physiological and pathological endogenous Ouabain levels in humans. P-CTX-3C cytotoxic effect, on human neurons co-incubated with OV (Ouabain+Veratridine) mix, was expressed starting from 100pM after short- and 25pM after long-term exposure. Notably, P-CTX-3C alone at 25nM induced cytotoxicity after 24h and prolonged exposure. This human brain-derived cell line appears a suitable cell-based-model to evaluate cytotoxicity of CTX present in marine food contaminated at low toxic levels and to characterize the toxicological profile of other/new congeners.
Collapse
Affiliation(s)
- Teresa Coccini
- Laboratory of Clinical and Experimental Toxicology, Poison Control Centre, Toxicology Unit, Maugeri Clinical Scientific Institutes S.p.A.-BS, IRCCS Pavia, Pavia Italy.
| | - Francesca Caloni
- Department of Veterinary Medicine (DIMEVET), Università degli Studi di Milano, Milano, Italy
| | - Uliana De Simone
- Laboratory of Clinical and Experimental Toxicology, Poison Control Centre, Toxicology Unit, Maugeri Clinical Scientific Institutes S.p.A.-BS, IRCCS Pavia, Pavia Italy
| |
Collapse
|
24
|
On the Many Actions of Ouabain: Pro-Cystogenic Effects in Autosomal Dominant Polycystic Kidney Disease. Molecules 2017; 22:molecules22050729. [PMID: 28467389 PMCID: PMC5688955 DOI: 10.3390/molecules22050729] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/19/2017] [Accepted: 04/30/2017] [Indexed: 02/07/2023] Open
Abstract
Ouabain and other cardenolides are steroidal compounds originally discovered in plants. Cardenolides were first used as poisons, but after finding their beneficial cardiotonic effects, they were rapidly included in the medical pharmacopeia. The use of cardenolides to treat congestive heart failure remained empirical for centuries and only relatively recently, their mechanisms of action became better understood. A breakthrough came with the discovery that ouabain and other cardenolides exist as endogenous compounds that circulate in the bloodstream of mammals. This elevated these compounds to the category of hormones and opened new lines of investigation directed to further study their biological role. Another important discovery was the finding that the effect of ouabain was mediated not only by inhibition of the activity of the Na,K-ATPase (NKA), but by the unexpected role of NKA as a receptor and a signal transducer, which activates a complex cascade of intracellular second messengers in the cell. This broadened the interest for ouabain and showed that it exerts actions that go beyond its cardiotonic effect. It is now clear that ouabain regulates multiple cell functions, including cell proliferation and hypertrophy, apoptosis, cell adhesion, cell migration, and cell metabolism in a cell and tissue type specific manner. This review article focuses on the cardenolide ouabain and discusses its various in vitro and in vivo effects, its role as an endogenous compound, its mechanisms of action, and its potential use as a therapeutic agent; placing especial emphasis on our findings of ouabain as a pro-cystogenic agent in autosomal dominant polycystic kidney disease (ADPKD).
Collapse
|
25
|
Ketchem CJ, Conner CD, Murray RD, DuPlessis M, Lederer ED, Wilkey D, Merchant M, Khundmiri SJ. Low dose ouabain stimulates NaK ATPase α1 subunit association with angiotensin II type 1 receptor in renal proximal tubule cells. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1863:2624-2636. [PMID: 27496272 PMCID: PMC5206903 DOI: 10.1016/j.bbamcr.2016.07.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/15/2016] [Accepted: 07/29/2016] [Indexed: 01/25/2023]
Abstract
Our laboratory has recently demonstrated that low concentrations of ouabain increase blood pressure in rats associated with stimulation of NaK ATPase activity and activation of the Src signaling cascade in NHE1-dependent manner. Proteomic analysis of human kidney proximal tubule cells (HKC11) suggested that the Angiotensin II type 1 receptor (AT1R) as an ouabain-associating protein. We hypothesize that ouabain-induced stimulation of NaK ATPase activity is mediated through AT1R. To test this hypothesis, we examined the effect of ouabain on renal cell angiotensin II production, the effect of AT1R inhibition on ouabain-stimulated NKA activity, and the effect of ouabain on NKA-AT1R association. Ouabain increased plasma angiotensin II levels in rats treated with ouabain (1μg/kg body wt./day) for 9days and increased angiotensin II levels in cell culture media after 24h treatment with ouabain in human (HKC11), mouse (MRPT), and human adrenal cells. Ouabain 10pM stimulated NKA-mediated 86Rb uptake and phosphorylation of EGFR, Src, and ERK1/2. These effects were prevented by the AT1R receptor blocker candesartan. FRET and TIRF microscopy using Bodipy-labeled ouabain and mCherry-NKA or mCherry-AT1R demonstrated association of ouabain with AT1R and NKA. Further our FRET and TIRF studies demonstrated increased association between AT1R and NKA upon treatment with low dose ouabain. We conclude that ouabain stimulates NKA in renal proximal tubule cells through an angiotensin/AT1R-dependent mechanism and that this pathway contributes to cardiac glycoside associated hypertension.
Collapse
Affiliation(s)
| | | | - Rebecca D Murray
- Department of Physiology, University of Louisville, KY, USA; Department of Medicine, University of Louisville, KY, USA
| | | | - Eleanor D Lederer
- Department of Physiology, University of Louisville, KY, USA; Department of Medicine, University of Louisville, KY, USA; Robley Rex VA Medical Center, Louisville, KY, USA
| | - Daniel Wilkey
- Department of Medicine, University of Louisville, KY, USA
| | | | - Syed J Khundmiri
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC, USA.
| |
Collapse
|
26
|
Iatrino R, Manunta P, Zagato L. Salt Sensitivity: Challenging and Controversial Phenotype of Primary Hypertension. Curr Hypertens Rep 2016; 18:70. [DOI: 10.1007/s11906-016-0677-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
27
|
Abstract
During the past 20 years, the studies on genetics or pharmacogenomics of primary hypertension provided interesting results supporting the role of genetics, but no actionable finding ready to be translated into personalized medicine. Two types of approaches have been applied: a "hypothesis-driven" approach on the candidate genes, coding for proteins involved in the biochemical machinery underlying the regulation of BP, and an "unbiased hypothesis-free" approach with GWAS, based on the randomness principles of frequentist statistics. During the past 10-15 years, the application of the latter has overtaken the application of the former leading to an enlargement of the number of previously unknown candidate loci or genes but without any actionable result for the therapy of hypertension. In the present review, we summarize the results of our hypothesis-driven approach based on studies carried out in rats with genetic hypertension and in humans with essential hypertension at the pre-hypertensive and early hypertensive stages. These studies led to the identification of mutant adducin and endogenous ouabain as candidate genetic-molecular mechanisms in both species. Rostafuroxin has been developed for its ability to selectively correct Na(+) pump abnormalities sustained by the two abovementioned mechanisms and to selectively reduce BP in rats and in humans carrying the gene variants underlying the mutant adducin and endogenous ouabain (EO) effects. A clinical trial is ongoing to substantiate these findings. Future studies should apply both the candidate gene and GWAS approaches to fully exploit the potential of genetics in optimizing the personalized therapy.
Collapse
|
28
|
Affiliation(s)
- John M Hamlyn
- From the Departments of Physiology (J.M.H., M.P.B.) and Medicine (M.P.B.), University of Maryland School of Medicine, Baltimore.
| | - Mordecai P Blaustein
- From the Departments of Physiology (J.M.H., M.P.B.) and Medicine (M.P.B.), University of Maryland School of Medicine, Baltimore.
| |
Collapse
|
29
|
Lanzani C, Gatti G, Citterio L, Messaggio E, Delli Carpini S, Simonini M, Casamassima N, Zagato L, Brioni E, Hamlyn JM, Manunta P. Lanosterol Synthase Gene Polymorphisms and Changes in Endogenous Ouabain in the Response to Low Sodium Intake. Hypertension 2015; 67:342-8. [PMID: 26667413 DOI: 10.1161/hypertensionaha.115.06415] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/18/2015] [Indexed: 11/16/2022]
Abstract
Circulating levels of endogenous ouabain (EO), a vasopressor hormone of adrenocortical origin, are increased by sodium depletion. Furthermore, lanosterol synthase, an enzyme involved in cholesterol biosynthesis, has a missense polymorphism (rs2254524 V642L) that affects EO biosynthesis in adrenocortical cells. Here, we investigated the hypothesis that lanosterol synthase rs2254524 alleles in vivo impact the blood pressure (BP) and EO responses evoked by a low dietary Na intake (<100 mEq/d, 2 weeks) among patients with mild essential hypertension. During the low salt diet, the declines in both systolic BP (SBP: -8.7±1.7 versus -3.0±1.5; P=0.013) and diastolic BP (DBP: -5.1±0.98 versus -1.4±0.94 mm Hg; P<0.05), and the slope of the long-term pressure-natriuresis relationship affected significantly the presence of the lanosterol synthase rs2254524 A variant (AA: 0.71±0.22, AC 0.09±0.13, and CC 0.04±0.11 mEq/mm Hg/24 h; P=0.028). In addition, BP rose in ≈25% of the patients in response to the low salt diet and this was associated with increased circulating EO. Lanosterol synthase gene polymorphisms influence both the salt sensitivity of BP and changes in circulating EO in response to a low salt diet. The response of BP and EO to the low salt diet is markedly heterogeneous. Approximately 25% of patients experienced adverse effects, that is, increased BP and EO when salt intake was reduced and may be at increased long-term risk. The augmented response of EO to the low salt diet further supports the view that adrenocortical function is abnormal in some essential hypertensives.
Collapse
Affiliation(s)
- Chiara Lanzani
- From the Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milan, Italy (C.L., G.G., L.C., E.M., S.D.C., M.S., N.C., L.Z., E.B., P.M.); and Department of Physiology, University of Maryland, Baltimore (J.M.H.)
| | - Guido Gatti
- From the Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milan, Italy (C.L., G.G., L.C., E.M., S.D.C., M.S., N.C., L.Z., E.B., P.M.); and Department of Physiology, University of Maryland, Baltimore (J.M.H.)
| | - Lorena Citterio
- From the Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milan, Italy (C.L., G.G., L.C., E.M., S.D.C., M.S., N.C., L.Z., E.B., P.M.); and Department of Physiology, University of Maryland, Baltimore (J.M.H.)
| | - Elisabetta Messaggio
- From the Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milan, Italy (C.L., G.G., L.C., E.M., S.D.C., M.S., N.C., L.Z., E.B., P.M.); and Department of Physiology, University of Maryland, Baltimore (J.M.H.)
| | - Simona Delli Carpini
- From the Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milan, Italy (C.L., G.G., L.C., E.M., S.D.C., M.S., N.C., L.Z., E.B., P.M.); and Department of Physiology, University of Maryland, Baltimore (J.M.H.)
| | - Marco Simonini
- From the Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milan, Italy (C.L., G.G., L.C., E.M., S.D.C., M.S., N.C., L.Z., E.B., P.M.); and Department of Physiology, University of Maryland, Baltimore (J.M.H.)
| | - Nunzia Casamassima
- From the Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milan, Italy (C.L., G.G., L.C., E.M., S.D.C., M.S., N.C., L.Z., E.B., P.M.); and Department of Physiology, University of Maryland, Baltimore (J.M.H.)
| | - Laura Zagato
- From the Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milan, Italy (C.L., G.G., L.C., E.M., S.D.C., M.S., N.C., L.Z., E.B., P.M.); and Department of Physiology, University of Maryland, Baltimore (J.M.H.)
| | - Elena Brioni
- From the Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milan, Italy (C.L., G.G., L.C., E.M., S.D.C., M.S., N.C., L.Z., E.B., P.M.); and Department of Physiology, University of Maryland, Baltimore (J.M.H.)
| | - John M Hamlyn
- From the Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milan, Italy (C.L., G.G., L.C., E.M., S.D.C., M.S., N.C., L.Z., E.B., P.M.); and Department of Physiology, University of Maryland, Baltimore (J.M.H.)
| | - Paolo Manunta
- From the Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milan, Italy (C.L., G.G., L.C., E.M., S.D.C., M.S., N.C., L.Z., E.B., P.M.); and Department of Physiology, University of Maryland, Baltimore (J.M.H.).
| |
Collapse
|
30
|
Chen L, Song H, Wang Y, Lee JC, Kotlikoff MI, Pritchard TJ, Paul RJ, Zhang J, Blaustein MP. Arterial α2-Na+ pump expression influences blood pressure: lessons from novel, genetically engineered smooth muscle-specific α2 mice. Am J Physiol Heart Circ Physiol 2015. [PMID: 26209057 DOI: 10.1152/ajpheart.00430.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Arterial myocytes express α1-catalytic subunit isoform Na(+) pumps (75-80% of total), which are ouabain resistant in rodents, and high ouabain affinity α2-Na(+) pumps. Mice with globally reduced α2-pumps (but not α1-pumps), mice with mutant ouabain-resistant α2-pumps, and mice with a smooth muscle (SM)-specific α2-transgene (α2 (SM-Tg)) that induces overexpression all have altered blood pressure (BP) phenotypes. We generated α2 (SM-DN) mice with SM-specific α2 (not α1) reduction (>50%) using nonfunctional dominant negative (DN) α2. We compared α2 (SM-DN) and α2 (SM-Tg) mice to controls to determine how arterial SM α2-pumps affect vasoconstriction and BP. α2 (SM-DN) mice had elevated basal mean BP (mean BP by telemetry: 117 ± 4 vs. 106 ± 1 mmHg, n = 7/7, P < 0.01) and enhanced BP responses to chronic ANG II infusion (240 ng·kg(-1)·min(-1)) and high (6%) NaCl. Several arterial Ca(2+) transporters, including Na(+)/Ca(2+) exchanger 1 (NCX1) and sarcoplasmic reticulum and plasma membrane Ca(2+) pumps [sarco(endo)plasmic reticulum Ca(2+)-ATPase 2 (SERCA2) and plasma membrane Ca(2+)-ATPase 1 (PMCA1)], were also reduced (>50%). α2 (SM-DN) mouse isolated small arteries had reduced myogenic reactivity, perhaps because of reduced Ca(2+) transporter expression. In contrast, α2 (SM-Tg) mouse aortas overexpressed α2 (>2-fold), NCX1, SERCA2, and PMCA1 (43). α2 (SM-Tg) mice had reduced basal mean BP (104 ± 1 vs. 109 ± 2 mmHg, n = 15/9, P < 0.02) and attenuated BP responses to chronic ANG II (300-400 ng·kg(-1)·min(-1)) with or without 2% NaCl but normal myogenic reactivity. NCX1 expression was inversely related to basal BP in SM-α2 engineered mice but was directly related in SM-NCX1 engineered mice. NCX1, which usually mediates arterial Ca(2+) entry, and α2-Na(+) pumps colocalize at plasma membrane-sarcoplasmic reticulum junctions and functionally couple via the local Na(+) gradient to help regulate cell Ca(2+). Altered Ca(2+) transporter expression in SM-α2 engineered mice apparently compensates to minimize Ca(2+) overload (α2 (SM-DN)) or depletion (α2 (SM-Tg)) and attenuate BP changes. In contrast, Ca(2+) transporter upregulation, observed in many rodent hypertension models, should enhance Ca(2+) entry and signaling and contribute significantly to BP elevation.
Collapse
Affiliation(s)
- Ling Chen
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland; Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Hong Song
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Youhua Wang
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jane C Lee
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - Michael I Kotlikoff
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - Tracy J Pritchard
- College of Nursing, College of Medicine, University of Cincinnati, Cincinnati, Ohio; and
| | - Richard J Paul
- Department of Molecular and Cell Physiology, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Jin Zhang
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Mordecai P Blaustein
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland; Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland;
| |
Collapse
|
31
|
Cabinet of Curiosities: Venom Systems and Their Ecological Function in Mammals, with a Focus on Primates. Toxins (Basel) 2015; 7:2639-58. [PMID: 26193318 PMCID: PMC4516934 DOI: 10.3390/toxins7072639] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/01/2015] [Accepted: 07/10/2015] [Indexed: 11/17/2022] Open
Abstract
Venom delivery systems (VDS) are common in the animal kingdom, but rare amongst mammals. New definitions of venom allow us to reconsider its diversity amongst mammals by reviewing the VDS of Chiroptera, Eulipotyphla, Monotremata, and Primates. All orders use modified anterior dentition as the venom delivery apparatus, except Monotremata, which possesses a crural system. The venom gland in most taxa is a modified submaxillary salivary gland. In Primates, the saliva is activated when combined with brachial gland exudate. In Monotremata, the crural spur contains the venom duct. Venom functions include feeding, intraspecific competition, anti-predator defense and parasite defense. Including mammals in discussion of venom evolution could prove vital in our understanding protein functioning in mammals and provide a new avenue for biomedical and therapeutic applications and drug discovery.
Collapse
|
32
|
Hamlyn JM, Manunta P. Endogenous cardiotonic steroids in kidney failure: a review and an hypothesis. Adv Chronic Kidney Dis 2015; 22:232-44. [PMID: 25908473 DOI: 10.1053/j.ackd.2014.12.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 12/17/2014] [Accepted: 12/22/2014] [Indexed: 02/06/2023]
Abstract
In response to progressive nephron loss, volume and humoral signals in the circulation have increasing relevance. These signals, including plasma sodium, angiotensin II, and those related to volume status, activate a slow neuromodulatory pathway within the central nervous system (CNS). The slow CNS pathway includes specific receptors for angiotensin II, mineralocorticoids, and endogenous ouabain (EO). Stimulation of the pathway leads to elevated sympathetic nervous system activity (SNA) and increased circulating EO. The sustained elevation of circulating EO (or ouabain) stimulates central and peripheral mechanisms that amplify the impact of SNA on vascular tone. These include changes in synaptic plasticity in the brain and sympathetic ganglia that increase preganglionic tone and amplify ganglionic transmission, amplification of the impact of SNA on arterial tone in the vascular wall, and the reprogramming of calcium signaling proteins in arterial myocytes. These increase SNA, raise basal and evoked arterial tone, and elevate blood pressure (BP). In the setting of CKD, we suggest that sustained activation/elevation of the slow CNS pathway, plasma EO, and the cardiotonic steroid marinobufagenin, comprises a feed-forward system that raises BP and accelerates kidney and cardiac damage. Block of the slow CNS pathway and/or circulating EO and marinobufagenin may reduce BP and slow the progression to ESRD.
Collapse
|
33
|
Blaustein MP. Reply to "Letter to the editor: 'Why isn't clinical experience with ouabain more widely accepted?'". Am J Physiol Heart Circ Physiol 2014; 307:H1264-5. [PMID: 25320336 DOI: 10.1152/ajpheart.00571.2014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Mordecai P Blaustein
- Departments of Physiology and Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
34
|
Davel AP, Couto GK, Wenceslau CF, Peres EC, Xavier FE, Rossoni LV. Enhanced Na⁺, K⁺-ATPase activity and endothelial modulation decrease phenylephrine-induced contraction in aorta from ouabain-treated normotensive and hypertensive rats. Horm Mol Biol Clin Investig 2014; 18:113-22. [PMID: 25390007 DOI: 10.1515/hmbci-2013-0058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 11/20/2013] [Indexed: 11/15/2022]
Abstract
AIM The purpose of this study was to compare the effect of long-term ouabain treatment on the vascular reactivity and Na+, K+-ATPase activity of a conductance artery from normotensive and hypertensive rats. METHODS Male Wistar rats were treated with ouabain (~8.0 µg/day, subcutaneously) or vehicle for 5 and 20 weeks, and spontaneously hypertensive rats (SHRs) for 5 weeks. Vasoconstrictor response to phenylephrine (10-10 to 10-4 M) and relaxation curves to KCl (1-10 mM) were analyzed in thoracic aorta. The effects of endothelial removal, L-NAME (100 μM), and indomethacin (10 μM) were used to evaluate the endothelial, nitric oxide (NO), and cyclooxygenase (COX) modulation of phenylephrine response, respectively. Protein expression of endothelial and neuronal NO synthase (NOS) and COX-2 were also investigated. RESULTS The phenylephrine-induced contraction was reduced, whereas the relaxation to KCl was enhanced in the aorta of ouabain-treated Wistar rats and SHRs. In both strains, endothelial modulation of α-adrenergic response was enhanced, related to an increased NO and reduced COX-derived vasoconstrictor factor modulation. Aortas from 20-week ouabain-treated Wistar rats showed reduced COX-2 and enhanced eNOS protein expression. In SHRs, 5-week ouabain treatment reduced COX-2 and increased nNOS protein expression. CONCLUSIONS The results suggest that long-term ouabain treatment reduces the α-adrenergic response of aorta from normotensive rats and SHRs, associated with an increase of NO synthesis, reduced COX-2-derived vasoconstrictor factors, and enhanced ouabain-sensitive Na+, K+-ATPase activity. These aortic mechanisms could be adjustments to the elevated blood pressure induced by ouabain, even in the presence of preexisting hypertension.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/physiology
- Blood Pressure/drug effects
- Cyclooxygenase 2/metabolism
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/physiology
- Hypertension/metabolism
- Hypertension/physiopathology
- Male
- Muscle Contraction
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Nitric Oxide Synthase Type I/metabolism
- Nitric Oxide Synthase Type III/metabolism
- Ouabain/pharmacology
- Phenylephrine/pharmacology
- Rats, Inbred SHR
- Rats, Wistar
- Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors
- Sodium-Potassium-Exchanging ATPase/metabolism
- Species Specificity
- Vasoconstriction/drug effects
- Vasoconstrictor Agents/pharmacology
- Vasodilation/drug effects
Collapse
|
35
|
Ferrandi M, Molinari I, Rastaldi MP, Ferrari P, Bianchi G, Manunta P. Rostafuroxin protects from podocyte injury and proteinuria induced by adducin genetic variants and ouabain. J Pharmacol Exp Ther 2014; 351:278-87. [PMID: 25187430 DOI: 10.1124/jpet.114.217133] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Glomerulopathies are important causes of morbidity and mortality. Selective therapies that address the underlying mechanisms are still lacking. Recently, two mechanisms, mutant β-adducin and ouabain, have been found to be involved in glomerular podocytopathies and proteinuria through nephrin downregulation. The main purpose of the present study was to investigate whether rostafuroxin, a novel antihypertensive agent developed as a selective inhibitor of Src-SH2 interaction with mutant adducin- and ouabain-activated Na,K-ATPase, may protect podocytes from adducin- and ouabain-induced effects, thus representing a novel pharmacologic approach for the therapy of podocytopathies and proteinuria caused by the aforementioned mechanisms. To study the effect of rostafuroxin on podocyte protein changes and proteinuria, mice carrying mutant β-adducin and ouabain hypertensive rats were orally treated with 100 μg/kg per day rostafuroxin. Primary podocytes from congenic rats carrying mutant α-adducin or β-adducin (NB) from Milan hypertensive rats and normal rat podocytes incubated with 10(-9) M ouabain were cultured with 10(-9) M rostafuroxin. The results indicated that mutant β-adducin and ouabain caused podocyte nephrin loss and proteinuria in animal models. These alterations were reproduced in primary podocytes from NB rats and normal rats incubated with ouabain. Treatment of animals, or incubation of cultured podocytes with rostafuroxin, reverted mutant β-adducin- and ouabain-induced effects on nephrin protein expression and proteinuria. We conclude that rostafuroxin prevented podocyte lesions and proteinuria due to mutant β-adducin and ouabain in animal models. This suggests a potential therapeutic effect of rostafuroxin in patients with glomerular disease progression associated with these two mechanisms.
Collapse
Affiliation(s)
- Mara Ferrandi
- Prassis Sigma-Tau Research Institute, Settimo Milanese, Milan, Italy (M.F., I.M., P.F.); Division of Genetics and Cell Biology, Genomics of Renal Diseases and Hypertension Unit, IRCC San Raffaele Scientific Institute, Milan, Italy (M.F., I.M., P.M.); Renal Research Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, Milan, Italy (M.P.R.); CVie Therapeutics, Hong Kong, China (P.F., G.B.); and Chair of Nephrology, University Vita-Salute, Milan, Italy (G.B., P.M.)
| | - Isabella Molinari
- Prassis Sigma-Tau Research Institute, Settimo Milanese, Milan, Italy (M.F., I.M., P.F.); Division of Genetics and Cell Biology, Genomics of Renal Diseases and Hypertension Unit, IRCC San Raffaele Scientific Institute, Milan, Italy (M.F., I.M., P.M.); Renal Research Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, Milan, Italy (M.P.R.); CVie Therapeutics, Hong Kong, China (P.F., G.B.); and Chair of Nephrology, University Vita-Salute, Milan, Italy (G.B., P.M.)
| | - Maria Pia Rastaldi
- Prassis Sigma-Tau Research Institute, Settimo Milanese, Milan, Italy (M.F., I.M., P.F.); Division of Genetics and Cell Biology, Genomics of Renal Diseases and Hypertension Unit, IRCC San Raffaele Scientific Institute, Milan, Italy (M.F., I.M., P.M.); Renal Research Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, Milan, Italy (M.P.R.); CVie Therapeutics, Hong Kong, China (P.F., G.B.); and Chair of Nephrology, University Vita-Salute, Milan, Italy (G.B., P.M.)
| | - Patrizia Ferrari
- Prassis Sigma-Tau Research Institute, Settimo Milanese, Milan, Italy (M.F., I.M., P.F.); Division of Genetics and Cell Biology, Genomics of Renal Diseases and Hypertension Unit, IRCC San Raffaele Scientific Institute, Milan, Italy (M.F., I.M., P.M.); Renal Research Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, Milan, Italy (M.P.R.); CVie Therapeutics, Hong Kong, China (P.F., G.B.); and Chair of Nephrology, University Vita-Salute, Milan, Italy (G.B., P.M.)
| | - Giuseppe Bianchi
- Prassis Sigma-Tau Research Institute, Settimo Milanese, Milan, Italy (M.F., I.M., P.F.); Division of Genetics and Cell Biology, Genomics of Renal Diseases and Hypertension Unit, IRCC San Raffaele Scientific Institute, Milan, Italy (M.F., I.M., P.M.); Renal Research Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, Milan, Italy (M.P.R.); CVie Therapeutics, Hong Kong, China (P.F., G.B.); and Chair of Nephrology, University Vita-Salute, Milan, Italy (G.B., P.M.)
| | - Paolo Manunta
- Prassis Sigma-Tau Research Institute, Settimo Milanese, Milan, Italy (M.F., I.M., P.F.); Division of Genetics and Cell Biology, Genomics of Renal Diseases and Hypertension Unit, IRCC San Raffaele Scientific Institute, Milan, Italy (M.F., I.M., P.M.); Renal Research Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, Milan, Italy (M.P.R.); CVie Therapeutics, Hong Kong, China (P.F., G.B.); and Chair of Nephrology, University Vita-Salute, Milan, Italy (G.B., P.M.)
| |
Collapse
|
36
|
Menger L, Vacchelli E, Kepp O, Eggermont A, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Cardiac glycosides and cancer therapy. Oncoimmunology 2014; 2:e23082. [PMID: 23525565 PMCID: PMC3601180 DOI: 10.4161/onci.23082] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cardiac glycosides (CGs) are natural compounds sharing the ability to operate as potent inhibitors of the plasma membrane Na+/K+-ATPase, hence promoting—via an indirect mechanism—the intracellular accumulation of Ca2+ ions. In cardiomyocytes, increased intracellular Ca2+ concentrations exert prominent positive inotropic effects, that is, they increase myocardial contractility. Owing to this feature, two CGs, namely digoxin and digitoxin, have extensively been used in the past for the treatment of several cardiac conditions, including distinct types of arrhythmia as well as contractility disorders. Nowadays, digoxin is approved by the FDA and indicated for the treatment of congestive heart failure, atrial fibrillation and atrial flutter with rapid ventricular response, whereas the use of digitoxin has been discontinued in several Western countries. Recently, CGs have been suggested to exert potent antineoplastic effects, notably as they appear to increase the immunogenicity of dying cancer cells. In this Trial Watch, we summarize the mechanisms that underpin the unsuspected anticancer potential of CGs and discuss the progress of clinical studies that have evaluated/are evaluating the safety and efficacy of CGs for oncological indications.
Collapse
Affiliation(s)
- Laurie Menger
- Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France ; INSERM; U848; Villejuif, France
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Hamlyn JM, Linde CI, Gao J, Huang BS, Golovina VA, Blaustein MP, Leenen FHH. Neuroendocrine humoral and vascular components in the pressor pathway for brain angiotensin II: a new axis in long term blood pressure control. PLoS One 2014; 9:e108916. [PMID: 25275393 PMCID: PMC4183521 DOI: 10.1371/journal.pone.0108916] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 08/26/2014] [Indexed: 11/29/2022] Open
Abstract
Central nervous system (CNS) administration of angiotensin II (Ang II) raises blood pressure (BP). The rise in BP reflects increased sympathetic outflow and a slower neuromodulatory pressor mechanism mediated by CNS mineralocorticoid receptors (MR). We investigated the hypothesis that the sustained phase of hypertension is associated also with elevated circulating levels of endogenous ouabain (EO), and chronic stimulation of arterial calcium transport proteins including the sodium-calcium exchanger (NCX1), the type 6 canonical transient receptor potential protein (TRPC6), and the sarcoplasmic reticulum calcium ATPase (SERCA2). Wistar rats received a chronic intra-cerebroventricular infusion of vehicle (C) or Ang II (A, 2.5 ng/min, for 14 days) alone or combined with the MR blocker, eplerenone (A+E, 5 µg/day), or the aldosterone synthase inhibitor, FAD286 (A+F, 25 µg/day). Conscious mean BP increased (P<0.05) in A (123±4 mm Hg) vs all other groups. Blood, pituitary and adrenal samples were taken for EO radioimmunoassay (RIA), and aortas for NCX1, TRPC6 and SERCA2 immunoblotting. Central infusion of Ang II raised plasma EO (0.58±0.08 vs C 0.34±0.07 nM (P<0.05), but not in A + E and A + F groups as confirmed by off-line liquid chromatography (LC)-RIA and LC-multistage mass spectrometry. Two novel isomers of EO were elevated by Ang II; the second less polar isomer increased >50-fold in the A+F group. Central Ang II increased arterial expression of NCX1, TRPC6 and SERCA2 (2.6, 1.75 and 3.7-fold, respectively; P<0.01)) but not when co-infused with E or F. Adrenal and pituitary EO were unchanged. We conclude that brain Ang II activates a CNS-humoral axis involving plasma EO. The elevated EO reprograms peripheral ion transport pathways known to control arterial Na+ and Ca2+ homeostasis; this increases contractility and augments sympathetic effects. The new axis likely contributes to the chronic pressor effect of brain Ang II.
Collapse
Affiliation(s)
- John M. Hamlyn
- Department of Physiology, University of Maryland Baltimore, Baltimore, Maryland, United States of America
- * E-mail:
| | - Cristina I. Linde
- Department of Physiology, University of Maryland Baltimore, Baltimore, Maryland, United States of America
| | - Junjie Gao
- Department of Physiology, University of Maryland Baltimore, Baltimore, Maryland, United States of America
| | - Bing S. Huang
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Vera A. Golovina
- Department of Physiology, University of Maryland Baltimore, Baltimore, Maryland, United States of America
| | - Mordecai P. Blaustein
- Department of Physiology, University of Maryland Baltimore, Baltimore, Maryland, United States of America
- Department of Medicine, University of Maryland Baltimore, Baltimore, Maryland, United States of America
| | | |
Collapse
|
38
|
Lewis LK, Yandle TG, Hilton PJ, Jensen BP, Begg EJ, Nicholls MG. Endogenous ouabain is not ouabain. Hypertension 2014; 64:680-3. [PMID: 25001271 DOI: 10.1161/hypertensionaha.114.03919] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Lynley K Lewis
- From the Christchurch Heart Institute (L.K.L., T.G.Y.) and Department of Medicine (E.J.B., M.G.N.), University of Otago-Christchurch, Christchurch, New Zealand; Department of Medicine, St Thomas' Hospital, London, United Kingdom (P.J.H.); and Division of Toxicology, Canterbury Health Laboratories, Christchurch, New Zealand (B.P.J.)
| | - Timothy G Yandle
- From the Christchurch Heart Institute (L.K.L., T.G.Y.) and Department of Medicine (E.J.B., M.G.N.), University of Otago-Christchurch, Christchurch, New Zealand; Department of Medicine, St Thomas' Hospital, London, United Kingdom (P.J.H.); and Division of Toxicology, Canterbury Health Laboratories, Christchurch, New Zealand (B.P.J.)
| | - Philip J Hilton
- From the Christchurch Heart Institute (L.K.L., T.G.Y.) and Department of Medicine (E.J.B., M.G.N.), University of Otago-Christchurch, Christchurch, New Zealand; Department of Medicine, St Thomas' Hospital, London, United Kingdom (P.J.H.); and Division of Toxicology, Canterbury Health Laboratories, Christchurch, New Zealand (B.P.J.)
| | - Berit P Jensen
- From the Christchurch Heart Institute (L.K.L., T.G.Y.) and Department of Medicine (E.J.B., M.G.N.), University of Otago-Christchurch, Christchurch, New Zealand; Department of Medicine, St Thomas' Hospital, London, United Kingdom (P.J.H.); and Division of Toxicology, Canterbury Health Laboratories, Christchurch, New Zealand (B.P.J.)
| | - Evan J Begg
- From the Christchurch Heart Institute (L.K.L., T.G.Y.) and Department of Medicine (E.J.B., M.G.N.), University of Otago-Christchurch, Christchurch, New Zealand; Department of Medicine, St Thomas' Hospital, London, United Kingdom (P.J.H.); and Division of Toxicology, Canterbury Health Laboratories, Christchurch, New Zealand (B.P.J.)
| | - M Gary Nicholls
- From the Christchurch Heart Institute (L.K.L., T.G.Y.) and Department of Medicine (E.J.B., M.G.N.), University of Otago-Christchurch, Christchurch, New Zealand; Department of Medicine, St Thomas' Hospital, London, United Kingdom (P.J.H.); and Division of Toxicology, Canterbury Health Laboratories, Christchurch, New Zealand (B.P.J.).
| |
Collapse
|
39
|
Affiliation(s)
- Mordecai P Blaustein
- Departments of Physiology and Medicine and the Center for Heart, Hypertension and Kidney Disease, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
40
|
Simonini M, Lanzani C, Bignami E, Casamassima N, Frati E, Meroni R, Messaggio E, Alfieri O, Hamlyn J, Body SC, Collard CD, Zangrillo A, Manunta P. A new clinical multivariable model that predicts postoperative acute kidney injury: impact of endogenous ouabain. Nephrol Dial Transplant 2014; 29:1696-701. [PMID: 24920842 DOI: 10.1093/ndt/gfu200] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is an important complication of cardiac surgery. Recently, elevated levels of endogenous ouabain (EO), an adrenal stress hormone with haemodynamic and renal effects, have been associated with worse renal outcome after cardiac surgery. Our aim was to develop and evaluate a new risk model of AKI using simple preoperative clinical parameters and to investigate the utility of EO. METHODS The primary outcome was AKI according to Acute Kidney Injury Network stage II or III. We selected the Northern New England Cardiovascular Disease Study Group (NNECDSG) as a reference model. We built a new internal predictive risk model considering common clinical variables (CLIN-RISK), compared this model with the NNECDSG model and determined whether the addition of preoperative plasma EO improved prediction of AKI. RESULTS All models were tested on >800 patients admitted for elective cardiac surgery in our hospital. Seventy-nine patients developed AKI (9.9%). Preoperative EO levels were strongly associated with the incidence of AKI and clinical complication (total ICU stay and in-hospital mortality). The NNECDSG model was confirmed as a good predictor of AKI (AUC 0.74, comparable to the NNECDSG reference population). Our CLIN-RISK model had improved predictive power for AKI (AUC 0.79, CI 95% 0.73-0.84). Furthermore, addition of preoperative EO levels to both clinical models improved AUC to 0.79 and to 0.83, respectively (ΔAUC +0.05 and +0.04, respectively, P < 0.01). CONCLUSION In a population where the predictive power of the NNECDSG model was confirmed, CLIN-RISK was more powerful. Both clinical models were further improved by the addition of preoperative plasma EO levels. These new models provide improved predictability of the relative risk for the development of AKI following cardiac surgery and suggest that EO is a marker for renal vascular injury.
Collapse
Affiliation(s)
- Marco Simonini
- Chair of Nephrology, Università Vita Salute San Raffaele, Milan, Italy Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Lanzani
- Chair of Nephrology, Università Vita Salute San Raffaele, Milan, Italy Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elena Bignami
- Anesthesia and Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy Università Vita Salute San Raffaele, Milan, Italy
| | - Nunzia Casamassima
- Chair of Nephrology, Università Vita Salute San Raffaele, Milan, Italy Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elena Frati
- Anesthesia and Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy Università Vita Salute San Raffaele, Milan, Italy
| | - Roberta Meroni
- Anesthesia and Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy Università Vita Salute San Raffaele, Milan, Italy
| | - Elisabetta Messaggio
- Chair of Nephrology, Università Vita Salute San Raffaele, Milan, Italy Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ottavio Alfieri
- Università Vita Salute San Raffaele, Milan, Italy Cardiac Surgery Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - John Hamlyn
- Physiology Department, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Simon C Body
- Department of Anesthesiology, Perioperative & Pain Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - C David Collard
- Baylor St. Luke's Medical Center, Texas Heart Institute, Houston, TX, USA
| | - Alberto Zangrillo
- Anesthesia and Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy Università Vita Salute San Raffaele, Milan, Italy
| | - Paolo Manunta
- Chair of Nephrology, Università Vita Salute San Raffaele, Milan, Italy Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | |
Collapse
|
41
|
Ferrandi M, Molinari I, Matafora V, Zerbini G, Trevisani F, Rastaldi M, Simonini M, Giardino L, Ferrari P, Manunta P. RETRACTED: SIK1 localizes with nephrin in glomerular podocytes and its polymorphism predicts kidney injury. Hum Mol Genet 2014; 23:4371-82. [DOI: 10.1093/hmg/ddu154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
42
|
Hamlyn JM. Natriuretic hormones, endogenous ouabain, and related sodium transport inhibitors. Front Endocrinol (Lausanne) 2014; 5:199. [PMID: 25520702 PMCID: PMC4253959 DOI: 10.3389/fendo.2014.00199] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 11/10/2014] [Indexed: 01/08/2023] Open
Abstract
The work of deWardener and colleagues stimulated longstanding interest in natriuretic hormones (NHs). In addition to the atrial peptides (APs), the circulation contains unidentified physiologically relevant NHs. One NH is controlled by the central nervous system (CNS) and likely secreted by the pituitary. Its circulating activity is modulated by salt intake and the prevailing sodium concentration of the blood and intracerebroventricular fluid, and contributes to postprandial and dehydration natriuresis. The other NH, mobilized by atrial stretch, promotes natriuresis by increasing the production of intrarenal dopamine and/or nitric oxide (NO). Both NHs have short (<35 min) circulating half lives, depress renotubular sodium transport, and neither requires the renal nerves. The search for NHs led to endogenous cardiotonic steroids (CTS) including ouabain-, digoxin-, and bufadienolide-like materials. These CTS, given acutely in high nanomole to micromole amounts into the general or renal circulations, inhibit sodium pumps and are natriuretic. Among these CTS, only bufalin is cleared sufficiently rapidly to qualify for an NH-like role. Ouabain-like CTS are cleared slowly, and when given chronically in low daily nanomole amounts, promote sodium retention, augment arterial myogenic tone, reduce renal blood flow and glomerular filtration, suppress NO in the renal vasa recta, and increase sympathetic nerve activity and blood pressure. Moreover, lowering total body sodium raises circulating endogenous ouabain. Thus, ouabain-like CTS have physiological actions that, like aldosterone, support renal sodium retention and blood pressure. In conclusion, the mammalian circulation contains two non-AP NHs. Identification of the CNS NH should be a priority.
Collapse
Affiliation(s)
- John M. Hamlyn
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- *Correspondence: John M. Hamlyn, Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA e-mail:
| |
Collapse
|
43
|
Liu M, Li Y, Citterio L, Huang QF, Zeng WF, Sheng CS, Wei FF, Dong Q, Li GL, Kang YY, Zhang L, Xu TY, Li JJ, Song J, Manunta P, Wang JG. A functional common polymorphism of the ABCB1 gene is associated with chronic kidney disease and hypertension in Chinese. Am J Hypertens 2013; 26:1428-36. [PMID: 23926124 DOI: 10.1093/ajh/hpt126] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Permeability glycoprotein is encoded by the ATP-binding cassette B1 gene (ABCB1) and is an extruder of toxic metabolites in the kidney. A functional common polymorphism (C3435T, rs1045642) in the human ABCB1 gene has been found to be associated with allograft outcome in kidney transplant patients. In this study, we investigated the association of the C3435T polymorphism with renal function and blood pressure (BP) in 2 Chinese populations. METHODS The discovery and replication populations were recruited from a mountainous area (Zhejiang Province) and a newly urbanized suburban area (Shanghai), respectively. We genotyped all subjects using the ABI SNapShot method. Chronic kidney disease (CKD) was defined as an estimated glomerular filtration rate <60 ml/min × 1.73 m(2) or 24-hour urinary albumin excretion ≥30 mg. RESULTS In the discovery population of 1,987 subjects, after adjustment for covariables, TT homozygosity (n = 217) was associated with a higher risk of CKD (n = 369; odds ratio (OR) = 1.73; P = 0.003) and with higher systolic BP (+3.1 mm Hg; P = 0.03) and pulse pressure (+3.4 mm Hg; P = 0.001). These associations were dependent on age (Pint ≤ 0.05). In subjects aged ≥60 years (n = 374), the corresponding OR or difference was 2.40 for CKD, 15.1 mm Hg for systolic BP, and 12.4 mm Hg for pulse pressure (P < 0.001). In similar adjusted analyses in the replication population of 2,427 elderly (≥60 years) subjects, TT homozygosity was also associated with a higher risk of CKD (OR = 1.39; P = 0.02) and an enhanced association of hypertension with CKD (OR = 1.50; P = 0.04). CONCLUSIONS The ABCB1 C3435T polymorphism might predict CKD, especially in the elderly.
Collapse
Affiliation(s)
- Ming Liu
- Centre for Epidemiological Studies and Clinical Trials, Shanghai Key Lab of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Xie JX, Li X, Xie Z. Regulation of renal function and structure by the signaling Na/K-ATPase. IUBMB Life 2013; 65:991-8. [PMID: 24323927 PMCID: PMC5375025 DOI: 10.1002/iub.1229] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 11/08/2013] [Indexed: 12/23/2022]
Abstract
The Na/K-ATPase as an essential ion pump was discovered more than 50 years ago (Skou (1989) Biochim. Biophys. Acta 1000, 439-446; Feraille and Doucet (2001) Physiol. Rev. 81, 345-418). The signaling function of Na/K-ATPase has been gradually appreciated over the last 20 years, first from the studies of regulatory effects of ouabain on cardiac cell growth. Several reviews on this topic have been written during the last few years (Schoner and Scheiner-Bobis (2007) Am. J. Physiol. Cell. Physiol. 293, C509-C536; Xie and Cai (2003) Mol. Interv. 3, 157 - 168; Bagrov et al. (2009) Pharmacol. Rev. 61, 9-38; Tian and Xie (2008) Physiology 23, 205-211; Fontana et al. (2013) FEBS J. 280, 5450-5455; Blanco and Wallace (2013) Am. J. Physiol. Renal Physiol. 305, F797-F812). This article will focus on the molecular mechanism of Na/K-ATPase-mediated signal transduction and its potential regulatory role in renal physiology and diseases.
Collapse
Affiliation(s)
- Jeffrey X Xie
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, USA
| | | | | |
Collapse
|
45
|
Seale AP, Yamaguchi Y, Johnstone WM, Borski RJ, Lerner DT, Grau EG. Endocrine regulation of prolactin cell function and modulation of osmoreception in the Mozambique tilapia. Gen Comp Endocrinol 2013; 192:191-203. [PMID: 23722201 DOI: 10.1016/j.ygcen.2013.05.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 05/11/2013] [Accepted: 05/14/2013] [Indexed: 01/06/2023]
Abstract
Prolactin (PRL) cells of the Mozambique tilapia, Oreochromis mossambicus, are osmoreceptors by virtue of their intrinsic osmosensitivity coupled with their ability to directly regulate hydromineral homeostasis through the actions of PRL. Layered upon this fundamental osmotic reflex is an array of endocrine control of PRL synthesis and secretion. Consistent with its role in fresh water (FW) osmoregulation, PRL release in tilapia increases as extracellular osmolality decreases. The hyposmotically-induced release of PRL can be enhanced or attenuated by a variety of hormones. Prolactin release has been shown to be stimulated by gonadotropin-releasing hormone (GnRH), 17-β-estradiol (E2), testosterone (T), thyrotropin-releasing hormone (TRH), atrial natriuretic peptide (ANP), brain-natriuretic peptide (BNP), C-type natriuretic peptide (CNP), ventricular natriuretic peptide (VNP), PRL-releasing peptide (PrRP), angiotensin II (ANG II), leptin, insulin-like growth factors (IGFs), ghrelin, and inhibited by somatostatin (SS), urotensin-II (U-II), dopamine, cortisol, ouabain and vasoactive intestinal peptide (VIP). This review is aimed at providing an overview of the hypothalamic and extra-hypothalamic hormones that regulate PRL release in euryhaline Mozambique tilapia, particularly in the context on how they may modulate osmoreception, and mediate the multifunctional actions of PRL. Also considered are the signal transduction pathways through which these secretagogues regulate PRL cell function.
Collapse
Affiliation(s)
- A P Seale
- Hawai'i Institute of Marine Biology, University of Hawaii, Kaneohe, HI 96744, USA.
| | | | | | | | | | | |
Collapse
|
46
|
REN YANPING, ZHANG MINGJUAN, ZHANG TING, HUANG RUOWEN. Effect of ouabain on myocardial remodeling in rats. Exp Ther Med 2013; 6:65-70. [PMID: 23935720 PMCID: PMC3735870 DOI: 10.3892/etm.2013.1098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 04/03/2013] [Indexed: 11/20/2022] Open
Abstract
The aim of this study was to investigate the effect of ouabain (EO) on myocardial remodeling. Twenty-two adult male Sprague-Dawley rats were randomly divided into two groups: the rats in the EO group (n=12) were intraperitoneally injected with EO daily and those in the control group (n=10) were injected with physiological saline daily. After 8 weeks the rats were sacrificed. The ultrastructural changes in the myocardium were observed. The expression levels of voltage-gated potassium channel 4.2 (KV4.2) were detected by real-time quantitative reverse transcription-polymerase chain reaction. The effects of EO on the myocardial action potential and transient potassium efflux (Ito) were measured by patch clamping. The systolic blood pressure (SBP) of 10 of the 12 rats in the EO group, designated as the EO-sensitive (OS) rats, began to increase from the fifth week of treatment and was significantly higher compared with that of the control group 6 weeks later (P<0.01). The remaining 2 rats in the EO group that presented no increase in SBP following 8 weeks of treatment (P>0.05) were designated as EO-resistant (OR) rats. Pathological ultrastructural changes were significant in the apical mid-myocardium of the OS rats. No significant differences in KV4.2 expression were observed among the OS, OR and control rats. The patch clamp results revealed that EO prolongs the action potential duration, reduces Ito and triggers the electrical remodeling of the myocardium. EO induces a blood pressure increase and triggers structural and electrical remodeling.
Collapse
|
47
|
Blanco G, Wallace DP. Novel role of ouabain as a cystogenic factor in autosomal dominant polycystic kidney disease. Am J Physiol Renal Physiol 2013; 305:F797-812. [PMID: 23761677 DOI: 10.1152/ajprenal.00248.2013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The classic role of the Na-K-ATPase is that of a primary active transporter that utilizes cell energy to establish and maintain transmembrane Na(+) and K(+) gradients to preserve cell osmotic stability, support cell excitability, and drive secondary active transport. Recent studies have revealed that Na-K-ATPase located within cholesterol-containing lipid rafts serves as a receptor for cardiotonic steroids, including ouabain. Traditionally, ouabain was viewed as a toxin produced only in plants, and it was used in relatively high concentrations to experimentally block the pumping action of the Na-K-ATPase. However, the new and unexpected role of the Na-K-ATPase as a signal transducer revealed a novel facet for ouabain in the regulation of a myriad of cell functions, including cell proliferation, hypertrophy, apoptosis, mobility, and metabolism. The seminal discovery that ouabain is endogenously produced in mammals and circulates in plasma has fueled the interest in this endogenous molecule as a potentially important hormone in normal physiology and disease. In this article, we review the role of the Na-K-ATPase as an ion transporter in the kidney, the experimental evidence for ouabain as a circulating hormone, the function of the Na-K-ATPase as a signal transducer that mediates ouabain's effects, and novel results for ouabain-induced Na-K-ATPase signaling in cystogenesis of autosomal dominant polycystic kidney disease.
Collapse
Affiliation(s)
- Gustavo Blanco
- Dept. of Molecular and Integrative Physiology, 3901 Rainbow Blvd., Kansas City, KS 66160.
| | | |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Endogenous cardiotonic steroids (CTS) exert long-term effects on salt and blood pressure homeostasis. Here we discuss recent observations on mechanisms of salt sensitivity that involve endogenous ouabain and novel pathways in the brain and discuss their possible relationship to arterial and renal function in hypertension. RECENT FINDINGS Chronic elevation of brain sodium promotes sustained hypertension mediated by central endogenous ouabain and the Na(+) pump α-2 catalytic subunit. The intermediary pressor mechanism in the brain involves aldosterone biosynthesis, activation of mineralocorticoid receptors and increased epithelial sodium channel activity. In the periphery, elevated plasma CTS raise contractility and blood pressure by augmentation of sympathetic nerve responses, increasing arterial Ca(2+) signaling and blunting nitric oxide production in the renal medulla and collecting ducts. SUMMARY Endogenous ouabain in the brain appears to play a critical role in salt sensitivity and hypertension. In the periphery, the J-shaped relationship of plasma endogenous ouabain in response to short-term changes in salt balance in humans raises the possibility that endogenous ouabain contributes to the increased risk of adverse cardiovascular events associated with both low and high salt intakes.
Collapse
|
49
|
Yatabe MS, Yatabe J, Takano K, Murakami Y, Sakuta R, Abe S, Sanada H, Kimura J, Watanabe T. Effects of a high-sodium diet on renal tubule Ca2+ transporter and claudin expression in Wistar-Kyoto rats. BMC Nephrol 2012. [PMID: 23199000 PMCID: PMC3538060 DOI: 10.1186/1471-2369-13-160] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Urinary Ca2+ excretion increases with dietary NaCl. NaCl-induced calciuria may be associated with hypertension, urinary stone formation and osteoporosis, but its mechanism and long-term effects are not fully understood. This study examined alterations in the expressions of renal Ca2+ transporters, channels and claudins upon salt loading to better understand the mechanism of salt-induced urinary Ca2+ loss. Methods Eight-week old Wistar-Kyoto rats were fed either 0.3% or 8% NaCl diet for 8 weeks. Renal cortical expressions of Na+/Ca2+ exchanger 1 (NCX1), Ca2+ pump (PCMA1b), Ca2+ channel (TRPV5), calbindin-D28k, and claudins (CLDN-2, -7, -8, -16 and −19) were analyzed by quantitative PCR, western blot and/or immunohistochemistry. Results Fractional excretion of Ca2+ increased 6.0 fold with high-salt diet. Renal cortical claudin-2 protein decreased by approximately 20% with decreased immunological staining on tissue sections. Claudin-16 and −19 expressions were not altered. Renal cortical TRPV5, calbindin-D28k and NCX1 expressions increased 1.6, 1.5 and 1.2 fold, respectively. Conclusions Chronic high-salt diet decreased claudin-2 protein and increased renal TRPV5, calbindin-D28k, and NCX1. Salt loading is known to reduce the proximal tubular reabsorption of both Na+ and Ca2+. The reduction in claudin-2 protein expression may be partly responsible for the reduced Ca2+ reabsorption in this segment. The concerted upregulation of more distal Ca2+-transporting molecules may be a physiological response to curtail the loss of Ca2+, although the magnitude of compensation does not seem adequate to bring the urinary Ca2+ excretion down to that of the normal-diet group.
Collapse
Affiliation(s)
- Midori Sasaki Yatabe
- Department of Pharmacology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Zulian A, Linde CI, Pulina MV, Baryshnikov SG, Papparella I, Hamlyn JM, Golovina VA. Activation of c-SRC underlies the differential effects of ouabain and digoxin on Ca(2+) signaling in arterial smooth muscle cells. Am J Physiol Cell Physiol 2012. [PMID: 23195071 DOI: 10.1152/ajpcell.00337.2012] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiotonic steroids (CTS) of the strophanthus and digitalis families have opposing effects on long-term blood pressure (BP). This implies hitherto unrecognized divergent signaling pathways for these CTS. Prolonged ouabain treatment upregulates Ca(2+) entry via Na(+)/Ca(2+) exchanger-1 (NCX1) and TRPC6 gene-encoded receptor-operated channels in mesenteric artery smooth muscle cells (ASMCs) in vivo and in vitro. Here, we test the effects of digoxin on Ca(2+) entry and signaling in ASMC. In contrast to ouabain treatment, the in vivo administration of digoxin (30 μg·kg(-1)·day(-1) for 3 wk) did not raise BP and had no effect on resting cytolic free Ca(2+) concentration ([Ca(2+)](cyt)) or phenylephrine-induced Ca(2+) signals in isolated ASMCs. Expression of transporters in the α2 Na(+) pump-NCX1-TRPC6 Ca(2+) signaling pathway was not altered in arteries from digoxin-treated rats. Upregulated α2 Na(+) pumps and a phosphorylated form of the c-SRC protein kinase (pY419-Src, ~4.5-fold) were observed in ASMCs from rats treated with ouabain but not digoxin. Moreover, in primary cultured ASMCs from normal rats, treatment with digoxin (100 nM, 72 h) did not upregulate NCX1 and TRPC6 but blocked the ouabain-induced upregulation of these transporters. Pretreatment of ASMCs with the c-Src inhibitor PP2 (1 μM; 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine) but not its inactive analog eliminated the effect of ouabain on NCX1 and TRPC6 expression and ATP-induced Ca(2+) entry. Thus, in contrast to ouabain, the interaction of digoxin with α2 Na(+) pumps is unable to activate c-Src phosphorylation and upregulate the downstream NCX1-TRPC6 Ca(2+) signaling pathway in ASMCs. The inability of digoxin to upregulate c-Src may underlie its inability to raise long-term BP.
Collapse
Affiliation(s)
- Alessandra Zulian
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | | | |
Collapse
|