Ponad 7000 publikacji medycznych!
Statystyki za 2021 rok:
odsłony: 8 805 378
Artykuły w Czytelni Medycznej o SARS-CoV-2/Covid-19

Poniżej zamieściliśmy fragment artykułu. Informacja nt. dostępu do pełnej treści artykułu
© Borgis - Postępy Nauk Medycznych 9/2016, s. 653-658
Katarzyna Łabno-Kirszniok, Grzegorz Piecha, *Andrzej Więcek
From salt sensitivity to hypertension – what do we know about endogenous cardiotonic steroids?**
Od sodowrażliwości do nadciśnienia tętniczego – co wiemy na temat endogennych steroidów kardiotonicznych?
Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia in Katowice
Head of Department: Professor Andrzej Więcek, MD, PhD
Streszczenie
Nadmierna podaż sodu w diecie jest jednym z głównych czynników sprzyjających rozwojowi nadciśnienia tętniczego. Endogenne steroidy kardiotoniczne (CTS) wydają się pełnić znaczącą rolę w patogenezie sodowrażliwego nadciśnienia tętniczego. Związki te pełnią rolę specyficznych cząsteczek sygnałowych (ligandów), mających zdolność wiązania się z pompą sodowo-potasową (Na/K ATPazą), jednym z ważniejszych enzymów białkowych odpowiedzialnych za przezbłonowy transport jonów sodu i potasu. Endogenne steroidy kardiotoniczne nie tylko hamują aktywność Na/K ATPazy, ale także uczestniczą w aktywacji szlaków przekaźnictwa wewnątrzkomórkowego. Mechanizmy te leżą u podstaw licznych procesów komórkowych, ale także, w odpowiedzi na nadmierne spożycie soli, prowadzą do nieprawidłowego nerkowego wydalania sodu oraz obkurczenia naczyń. Istnieje bezpośrednia zależność pomiędzy stężeniem krążących steroidów kardiotonicznych w osoczu a średnim ciśnieniem tętniczym u ludzi. Ponadto, ostatnie badania sugerują udział endogennych steroidów kardiotonicznych w patogenezie włóknienia w układzie sercowo-naczyniowym i nerkach.
Summary
Dietary sodium intake is one of the major risk factors for developing high blood pressure. Endogenous cardiotonic steroids (CTS) have been proposed to play a significant role in salt-sensitive hypertension. They are a class of specific ligands for one of the most important membrane proteins, Na/K ATPase that play a central role in salt and water transport. Binding of cardiotonic steroids to the NA/K ATPase not only inhibits the enzyme activity but also activates intracellular signaling pathways. These mechanisms are known to underlie a number of cellular functions, but also contribute to impaired renal sodium excretion and vasoconstriction in response to an excessive dietary salt intake. A significant relationship has been shown between circulating levels of CTS and mean arterial pressure in humans. Moreover, recent studies implicates these hormones in the pathogenesis of cardiovascular and renal fibrosis.



Introduction
Dietary salt intake is an important factor implicated in the pathogenesis of hypertension. Both epidemiologic and observational studies have provided evidence that dietary sodium intake as well as urinary sodium excretion are closely associated with the prevalence of hypertension (1-3). There is also strong evidence that reduction in sodium intake can decrease blood pressure (4). In 1997, the Dietary Approaches to Stop Hypertension (DASH) trial has proven, that a low-sodium diet, rich in fruits and vegetables, can both prevent and treat hypertension (5). It has been demonstrated by Weinberger that increased salt loading causes an increase in blood pressure in all individuals (6). However, there is a substantial heterogeneity among-individuals in blood pressure responses to alterations in sodium and extracellular volume balance. The magnitude of this salt sensitivity is associated with a variety of demographic, physiological and genetic characteristics. It is possible to identify two main groups in the general population: salt-sensitive and salt-resistant. A variety of techniques and criteria have been proposed to assess the blood pressure response to changes in sodium and extracellular fluid balance, including specific maneuvers, such as intravenous infusion of saline, rapid sodium and volume depletion by diuretic administration or longer periods of dietary sodium manipulation. The largest epidemiological study conducted so far with 378 normotensive volunteers and 198 patients with essential hypertension, found 26% normotensive and 51% hypertensive subjects to be salt-sensitive (6).
Kidney and salt-sensitive hypertension
The cardiovascular system and the kidneys play an indispensable role in the regulation of arterial blood pressure. The kidneys play a central role in both development and maintenance of arterial hypertension through a direct control of sodium and water homeostasis. Evidence from a variety of studies in humans suggests an abnormality in salt handling by the kidneys as an underlying factor causing salt-sensitive hypertension (7-10). These alterations in renal function, may contribute to the etiology of salt-sensitive hypertension and are mediated by both genetic and environmental factors (11). Experimental studies have clearly shown that the central nervous system, alongside kidneys, plays a critical role in many forms of salt-sensitive hypertension (12-16).
Molecular mechanisms linking salt intake and blood pressure elevation are complex, multifactorial and remain unresolved. Recent studies have demonstrated that endogenous cardiotonic steroids (CTS) are important regulators of renal sodium excretion as well as blood pressure and may play a key role in the pathogenesis of salt-induced hypertension (17-19).
From a concept of natriuretic hormone to first CTS identification
The concept of circulating “humoral factor” hypothesized to induce salt-sensitive hypertension, came from the study performed by Dahl et al. in 1969 (20). In 1974, it was shown this pro-hypertensive “humoral factor” reduced activity of the sodium pump, the Na/K ATPase (21). A relationship between circulating Na/K ATPase inhibitors and blood pressure was first identified in humans in 1982. Moreover, there is a significant correlation between the level of these circulating inhibitors and mean arterial pressure in hypertensive patients (22). Based on numerous clinical studies and observations in volume expanded experimental animals, de Wardener and Clarkson suggested that a mysterious “humoral factor” implicated in pathogenesis of salt-sensitive hypertension is an endogenous natriuretic (23). Subsequent studies have shown that this presumptive “natriuretic hormone” has digitalis-like properties. Digitalis glycosides are specific ligands of Na/K ATPase and Na/K ATPase plays a major role in the renal tubular sodium transport. Therefore, a hypothesis was proposed that the essential role of endogenous digitalis is to promote natriuresis via inhibition of Na/K ATPase and sodium reabsorption in the renal proximal tubules (23). Moreover, endogenous digitalis-like factors could also contribute to vasoconstriction via inhibition of Na/K ATPase activity in vascular smooth muscle cells (24). In 1991 Hamlyn et al. suggested that their highly concentrated samples comprised a compound indistinguishable from plant-derived ouabain. Thus, endogenous ouabain (EO) was the first CTS to be identified in human plasma (25). Subsequent works by Bagrov et al. in patients after acute myocardial infarction have identified another widely studied member of the endogenous cardiotonic steroids, marinobufagenin (MBG) (26). Shortly thereafter, MBG has been detected in human plasma (27, 28).
Endogenous cardiotonic steroids (CTS) and Na/K ATPase
Cardiotonic steroids were first found in plants, most notably digitalis in the foxglove plant, and then in the skin of toads like the Bufo marinus (29). They have been used in traditional ancient medicine to treat congestive heart failure (30). CTS, or digitalis-like substances, are divided into two distinct groups related by structure: cardenolides, represented by digoxin and ouabain, and bufadienolides, represented by marinobufagenin and telocinobufagin. Bufadienolides differ from cardenolides in having a double-unsaturated six-atom lactone ring. Considering that, the amphibian skin participates in water and electrolyte homeostasis and the concentration of bufadienolides in toad skin is regulated by the salt content and its environment, it was hypothesized that the sodium pump (Na/K ATPase) and bufadienolides work together as a regulatory system serving as a basic stimulus-response coupling mechanism to maintain water and electrolyte balance (31, 32). Extremely important contributions were made by several groups which indicated that human fluids contain material that cross-react with antibodies against one of the bufadienolides, bufalin (33, 34). In the 1990s, endogenous ouabain and marinobufagenin were purified from human plasma.
Na/K ATPase is an active transport mechanism moving sodium and potassium ions across the cell membrane. This process is responsible for maintaining both an electrical and chemical gradient that is essential for maintaining a number of vital cell functions, such as communication, excitation, muscle contraction, and many other cellular functions. Na/K ATPase is a membrane-spanning enzyme expressed in virtually all cells of higher organisms. It structurally consists of two subunits, a large catalytic subunit α and a smaller glycoprotein subunit β (35).
Endogenous cardiotonic steroids bind to a specific site within the α subunit of Na/K ATPase and inhibit its activity. There are four isoforms of the α-subunit that have been identified in various tissues (36). The α1 isoform is ubiquitous and it is the main isoenzyme expressed in the renotubular epithelium (35). The α2 isoform is predominant in heart, vascular smooth muscle, skeletal muscle, adipocytes and brain. The α3 isoform is mostly found in neurons and in the cardiac tissue. The α4 isoform is expressed in the testis (37). The renal α1 isoform of Na/K ATPase plays a key role in the sodium reabsorption along the nephron. The α1 and α2 isoforms are a major determinant of smooth muscle contraction and vasoconstriction.

Powyżej zamieściliśmy fragment artykułu, do którego możesz uzyskać pełny dostęp.
Mam kod dostępu
  • Aby uzyskać płatny dostęp do pełnej treści powyższego artykułu albo wszystkich artykułów (w zależności od wybranej opcji), należy wprowadzić kod.
  • Wprowadzając kod, akceptują Państwo treść Regulaminu oraz potwierdzają zapoznanie się z nim.
  • Aby kupić kod proszę skorzystać z jednej z poniższych opcji.

Opcja #1

24

Wybieram
  • dostęp do tego artykułu
  • dostęp na 7 dni

uzyskany kod musi być wprowadzony na stronie artykułu, do którego został wykupiony

Opcja #2

59

Wybieram
  • dostęp do tego i pozostałych ponad 7000 artykułów
  • dostęp na 30 dni
  • najpopularniejsza opcja

Opcja #3

119

Wybieram
  • dostęp do tego i pozostałych ponad 7000 artykułów
  • dostęp na 90 dni
  • oszczędzasz 28 zł
Piśmiennictwo
Intersalt: an international study of electrolyte excretion and blood pressure. Results for 24 hour urinary sodium and potassium excretion. Intersalt Cooperative Research Group. BMJ 1988; 297(6644): 319-328.
Dahl LK: Possible role of chronic excess salt consumption in the pathogenesis of essential hypertension. Am J Cardiol 1961; 8: 571-575.
Yamori Y, Nara Y, Mizushima S et al.: International cooperative study on the relationship between dietary factors and blood pressure: a report from the Cardiovascular Diseases and Alimentary Comparison (CARDIAC) Study. J Cardiovasc Pharmacol 1990; 16 (suppl. 8): S43-47.
MacGregor GA, Markandu ND, Best FE et al.: Double-blind randomised crossover trial of moderate sodium restriction in essential hypertension. Lancet 1982; 1(8268): 351-355.
Appel LJ, Moore TJ, Obarzanek E et al.: A clinical trial of the effects of dietary patterns on blood pressure. DASH Collaborative Research Group. N Engl J Med 1997; 336(16): 1117-1124.
Weinberger MH: Salt sensitivity of blood pressure in humans. Hypertension 1996; 27(3 Pt 2): 481-490.
Parmer RJ, Stone RA, Cervenka JH: Renal hemodynamics in essential hypertension. Racial differences in response to changes in dietary sodium. Hypertension 1994; 24(6): 752-757.
Campese VM, Parise M, Karubian F, Bigazzi R: Abnormal renal hemodynamics in black salt-sensitive patients with hypertension. Hypertension 1991; 18(6): 805-812.
Wedler B, Brier ME, Wiersbitzky M et al.: Sodium kinetics in salt-sensitive and salt-resistant normotensive and hypertensive subjects. J Hypertens 1992; 10(7): 663-669.
Kimura G, Frem GJ, Brenner BM: Renal mechanisms of salt sensitivity in hypertension. Curr Opin Nephrol Hypertens 1994; 3(1): 1-12.
Folkow B: Physiological aspects of primary hypertension. Physiol Rev 1982; 62(2): 347-504.
Shah J, Jandhyala BS: Studies on the role(s) of cerebrospinal fluid osmolality and chloride ion in the centrally mediated pressor responses of sodium chloride. Clin Exp Hypertens A 1991; 13(2): 297-312.
Van Huysse JW, Amin MS, Yang B, Leenen FH: Salt-induced hypertension in a mouse model of Liddle syndrome is mediated by epithelial sodium channels in the brain. Hypertension 2012; 60(3): 691-696.
Reddy SR, Kotchen TA: Hemodynamic effects of high dietary intakes of sodium or chloride in the Dahl salt-sensitive rat. J Lab Clin Med 1992; 120(3): 476-482.
Leenen FH, Ruzicka M, Huang BS: The brain and salt-sensitive hypertension. Curr Hypertens Rep 2002; 4(2): 129-135.
Takahashi H: Upregulation of the Renin-Angiotensin-aldosterone-ouabain system in the brain is the core mechanism in the genesis of all types of hypertension. Int J Hypertens 2012; 2012: 242786.
Bagrov AY, Shapiro JI: Endogenous digitalis: pathophysiologic roles and therapeutic applications. Nat Clin Pract Nephrol 2008; 4(7): 378-392.
Blaustein MP, Leenen FH, Chen L et al.: How NaCl raises blood pressure: a new paradigm for the pathogenesis of salt-dependent hypertension. Am J Physiol Heart Circ Physiol 2012; 302(5): H1031-1049.
Bagrov AY, Fedorova OV: Cardenolide and bufadienolide ligands of the sodium pump. How they work together in NaCl sensitive hypertension. Front Biosci 2005; 10: 2250-2256.
Dahl LK, Knudsen KD, Iwai J: Humoral transmission of hypertension: evidence from parabiosis. Circ Res 1969; 24 (5 suppl.): 21-33.
Kramer HJ, Gonick HC: Effect of extracellular volume expansion on renal Na-K-ATPase and cell metabolism. Nephron 1974; 12(4): 281-296.
Hamlyn JM, Ringel R, Schaeffer J et al.: A circulating inhibitor of (Na+ + K+)ATPase associated with essential hypertension. Nature 1982; 300(5893): 650-652.
de Wardener HE, Clarkson EM: Concept of natriuretic hormone. Physiol Rev 1985; 65(3): 658-759.
Blaustein MP: Sodium ions, calcium ions, blood pressure regulation, and hypertension: a reassessment and a hypothesis. Am J Physiol 1977; 232(5): C165-173.
Hamlyn JM, Blaustein MP, Bova S et al.: Identification and characterization of a ouabain-like compound from human plasma. Proc Natl Acad Sci U S A 1991; 88(14): 6259-6263.
Bagrov AY, Fedorova OV, Dmitrieva RI et al.: Characterization of a urinary bufodienolide Na+,K+-ATPase inhibitor in patients after acute myocardial infarction. Hypertension 1998; 31(5): 1097-1103.
Fedorova OV, Doris PA, Bagrov AY: Endogenous marinobufagenin-like factor in acute plasma volume expansion. Clin Exp Hypertens 1998; 20(5-6): 581-591.
Gonick HC, Ding Y, Vaziri ND et al.: Simultaneous measurement of marinobufagenin, ouabain, and hypertension-associated protein in various disease states. Clin Exp Hypertens 1998; 20(5-6): 617-627.
Bagrov AY, Shapiro JI, Fedorova OV: Endogenous cardiotonic steroids: physiology, pharmacology, and novel therapeutic targets. Pharmacol Rev 2009; 61(1): 9-38.
Fisch C: William Withering: An account of the foxglove and some of its medical uses 1785-1985. J Am Coll Cardiol 1985; 5(5 suppl. A): 1A-2A.
Flier JS, Maratos-Flier E, Pallotta JA, McIsaac D: Endogenous digitalis-like activity in the plasma of the toad Bufo marinus. Nature 1979; 279(5711): 341-343.
Lichtstein D, Gati I, Haver E, Katz U: Digitalis-like compounds in the toad Bufo viridis: tissue and plasma levels and significance in osmotic stress. Life Sci 1992; 51(2): 119-128.
Goto A, Yamada K, Ishii M et al.: Immunoreactivity of endogenous digitalis-like factors. Biochem Pharmacol 1991; 41(8): 1261-1263.
Oda M, Kurosawa M, Numazawa S et al.: Determination of bufalin-like immunoreactivity in serum of humans and rats by time-resolved fluoroimmunoassay for using a monoclonal antibody. Life Sci 2001; 68(10): 1107-1117.
Skou JC, Esmann M: The Na,K-ATPase. J Bioenerg Biomembr 1992; 24(3): 249-261.
Lingrel JB, Kuntzweiler T: Na+,K(+)-ATPase. J Biol Chem 1994; 269(31): 19659-19662.
Dostanic-Larson I, Lorenz JN, Van Huysse JW et al.: Physiological role of the alpha1- and alpha2-isoforms of the Na+-K+-ATPase and biological significance of their cardiac glycoside binding site. Am J Physiol Regul Integr Comp Physiol 2006; 290(3): R524-528.
O’Brien WJ, Lingrel JB, Wallick ET: Ouabain binding kinetics of the rat alpha two and alpha three isoforms of the sodium-potassium adenosine triphosphate. Arch Biochem Biophys 1994; 310(1): 32-39.
Juhaszova M, Blaustein MP: Distinct distribution of different Na+ pump alpha subunit isoforms in plasmalemma. Physiological implications. Ann N Y Acad Sci 1997; 834: 524-536.
Iwamoto T: Vascular Na+/Ca2+ exchanger: implications for the pathogenesis and therapy of salt-dependent hypertension. Am J Physiol Regul Integr Comp Physiol 2006; 290(3): R536-545.
Zhang J, Ren C, Chen L et al.: Knockout of Na+/Ca2+ exchanger in smooth muscle attenuates vasoconstriction and L-type Ca2+ channel current and lowers blood pressure. Am J Physiol Heart Circ Physiol 2010; 298(5): H1472-1483.
Pulina MV, Zulian A, Berra-Romani R et al.: Upregulation of Na+ and Ca2+ transporters in arterial smooth muscle from ouabain-induced hypertensive rats. Am J Physiol Heart Circ Physiol 2010; 298(1): H263-274.
Wang J, Velotta JB, McDonough AA, Farley RA: All human Na(+)-K(+)-ATPase alpha-subunit isoforms have a similar affinity for cardiac glycosides. Am J Physiol Cell Physiol 2001; 281(4): C1336-1343.
Fedorova OV, Kolodkin NI, Agalakova NI et al.: Marinobufagenin, an endogenous alpha-1 sodium pump ligand, in hypertensive Dahl salt-sensitive rats. Hypertension 2001; 37(2 Pt 2): 462-466.
Roman RJ: Abnormal renal hemodynamics and pressure-natriuresis relationship in Dahl salt-sensitive rats. Am J Physiol 1986; 251(1 Pt 2): F57-65.
Fedorova OV, Lakatta EG, Bagrov AY: Endogenous Na,K pump ligands are differentially regulated during acute NaCl loading of Dahl rats. Circulation 2000; 102(24): 3009-3014.
Fedorova OV, Agalakova NI, Talan MI et al.: Brain ouabain stimulates peripheral marinobufagenin via angiotensin II signalling in NaCl-loaded Dahl-S rats. J Hypertens 2005; 23(8): 1515-1523.
Fedorova OV, Bagrov AY: Inhibition of Na/K ATPase from rat aorta by two Na/K pump inhibitors, ouabain and marinobufagenin: evidence of interaction with different alpha-subunit isoforms. Am J Hypertens 1997; 10(8): 929-935.
Fedorova OV, Talan MI, Agalakova NI et al.: Endogenous ligand of alpha(1) sodium pump, marinobufagenin, is a novel mediator of sodium chloride – dependent hypertension. Circulation 2002; 105(9): 1122-1127.
Bagrov AY, Agalakova NI, Kashkin VA, Fedorova OV: Endogenous cardiotonic steroids and differential patterns of sodium pump inhibition in NaCl-loaded salt-sensitive and normotensive rats. Am J Hypertens 2009; 22(5): 559-563.
Periyasamy SM, Liu J, Tanta F et al.: Salt loading induces redistribution of the plasmalemmal Na/K-ATPase in proximal tubule cells. Kidney Int 2005; 67(5): 1868-1877.
Oweis S, Wu L, Kiela PR et al.: Cardiac glycoside downregulates NHE3 activity and expression in LLC-PK1 cells. Am J Physiol Renal Physiol 2006; 290(5): F997-1008.
Liu J, Liang M, Liu L et al.: Ouabain-induced endocytosis of the plasmalemmal Na/K-ATPase in LLC-PK1 cells requires caveolin-1. Kidney Int 2005; 67(5): 1844-1854.
Li Z, Xie Z: The Na/K-ATPase/Src complex and cardiotonic steroid-activated protein kinase cascades. Pflugers Arch 2009; 457(3): 635-644.
Liu J, Xie ZJ: The sodium pump and cardiotonic steroids-induced signal transduction protein kinases and calcium-signaling microdomain in regulation of transporter trafficking. Biochim Biophys Acta 2010; 1802(12): 1237-1245.
Komiyama Y, Dong XH, Nishimura N et al.: A novel endogenous digitalis, telocinobufagin, exhibits elevated plasma levels in patients with terminal renal failure. Clin Biochem 2005; 38(1): 36-45.
Li S, Eim C, Kirch U et al.: Bovine adrenals and hypothalamus are a major source of proscillaridin A- and ouabain-immunoreactivities. Life Sci 1998; 62(11): 1023-1033.
Murrell JR, Randall JD, Rosoff J et al.: Endogenous ouabain: upregulation of steroidogenic genes in hypertensive hypothalamus but not adrenal. Circulation 2005; 112(9): 1301-1308.
el-Masri MA, Clark BJ, Qazzaz HM, Valdes R Jr: Human adrenal cells in culture produce both ouabain-like and dihydroouabain-like factors. Clin Chem 2002; 48(10): 1720-1730.
Fedorova OV, Zhuravin IA, Agalakova NI et al.: Intrahippocampal microinjection of an exquisitely low dose of ouabain mimics NaCl loading and stimulates a bufadienolide Na/K-ATPase inhibitor. J Hypertens 2007; 25(9): 1834-1844.
Anderson DE, Fedorova OV, Morrell CH et al.: Endogenous sodium pump inhibitors and age-associated increases in salt sensitivity of blood pressure in normotensives. Am J Physiol Regul Integr Comp Physiol 2008; 294(4): R1248-1254.
Manunta P, Hamilton BP, Hamlyn JM: Salt intake and depletion increase circulating levels of endogenous ouabain in normal men. Am J Physiol Regul Integr Comp Physiol 2006; 290(3): R553-559.
Staessen JA, Thijs L, Stolarz-Skrzypek K et al.: Main results of the ouabain and adducin for Specific Intervention on Sodium in Hypertension Trial (OASIS-HT): a randomized placebo-controlled phase-2 dose-finding study of rostafuroxin. Trials 2011; 12: 13.
Manunta P, Messaggio E, Ballabeni C et al.: Plasma ouabain-like factor during acute and chronic changes in sodium balance in essential hypertension. Hypertension 2001; 38(2): 198-203.
Jablonski KL, Fedorova OV, Racine ML et al.: Dietary sodium restriction and association with urinary marinobufagenin, blood pressure, and aortic stiffness. Clin J Am Soc Nephrol 2013; 8(11): 1952-1959.
Rossi G, Manunta P, Hamlyn JM et al.: Immunoreactive endogenous ouabain in primary aldosteronism and essential hypertension: relationship with plasma renin, aldosterone and blood pressure levels. J Hypertens 1995; 13(10): 1181-1191.
Tomaschitz A, Piecha G, Ritz E et al.: Marinobufagenin in essential hypertension and primary aldosteronism: a cardiotonic steroid with clinical and diagnostic implications. Clin Exp Hypertens 2015; 37(2): 108-115.
Elkareh J, Kennedy DJ, Yashaswi B et al.: Marinobufagenin stimulates fibroblast collagen production and causes fibrosis in experimental uremic cardiomyopathy. Hypertension 2007; 49(1): 215-224.
Fedorova LV, Raju V, El-Okdi N et al.: The cardiotonic steroid hormone marinobufagenin induces renal fibrosis: implication of epithelial-to-mesenchymal transition. Am J Physiol Renal Physiol 2009; 296(4): F922-934.
Briones AM, Xavier FE, Arribas SM et al.: Alterations in structure and mechanics of resistance arteries from ouabain-induced hypertensive rats. Am J Physiol Heart Circ Physiol 2006; 291(1): H193-201.
Pierdomenico SD, Bucci A, Manunta P et al.: Endogenous ouabain and hemodynamic and left ventricular geometric patterns in essential hypertension. Am J Hypertens 2001; 14(1): 44-50.
Ferrandi M, Molinari I, Barassi P et al.: Organ hypertrophic signaling within caveolae membrane subdomains triggered by ouabain and antagonized by PST 2238. J Biol Chem 2004; 279(32): 33306-33314.
Aydemir-Koksoy A, Abramowitz J, Allen JC: Ouabain-induced signaling and vascular smooth muscle cell proliferation. J Biol Chem 2001; 276(49): 46605-46611.
otrzymano: 2016-08-04
zaakceptowano do druku: 2016-08-25

Adres do korespondencji:
*Andrzej Więcek
Department of Nephrology, Transplantation and Internal Medicine Medical University of Silesia in Katowice
ul. Francuska 20-24, 40-027 Katowice
tel. +48 (32) 255-26-95
awiecek@sum.edu.pl

Postępy Nauk Medycznych 9/2016
Strona internetowa czasopisma Postępy Nauk Medycznych