Quick viewing(Text Mode)

A Dissertation Entitled Na/K-Atpase Mediates Renal Sodium Handling

A Dissertation Entitled Na/K-Atpase Mediates Renal Sodium Handling

A Dissertation

Entitled

Na/K-ATPase Mediates Renal Sodium Handling

By Yanling Yan

Submitted to the Graduate Faculty as partial fulfillment of the requirements for the

Doctor of Philosophy Degree in Biomedical Sciences

Dr. Joseph Shapiro, Committee Chair

Dr. Jiang Liu, Committee Member

Dr. Nader Abraham, Committee Member

Dr. Deepak Malhotra, Committee Member

Dr. Bina Joe, Committee Member

Dr. Zijian Xie, Committee Member

Dr. Patricia Komuniecki, Dean

College of Graduate Studies

The University of Toledo August 2012

Copyright 2012, Yanling Yan This document is copyrighted material. Under copyright law, no parts of this document may be reproduced without the expressed permission of the author.

An Abstract of

Na/K-ATPase Mediates Renal Sodium Handling

by

Yanling Yan

Submitted to the Graduate Faculty as partial fulfillment of the requirement for the Doctor of Philosophy Degree in Biomedical Sciences

The University of Toledo August 2012

Hypertension, affecting over one billion people worldwide, is one of the leading risk factors for heart attack and stroke. Kidney cross-transplantation studies between hypertensive and normotensive people and strains of rats provide the most compelling evidence for the fundamental role of the kidney in the pathogenesis of hypertension.

Recently, there is growing evidence supporting Arthur Guyton’s hypothesis that a

common characteristic feature of hypertension is impaired renal sodium excretion.

However, the exact molecular mechanism responsible for the impaired renal sodium

excretion is not clearly defined. The overall aim of this dissertation is to improve our

understanding of Na/K-ATPase and sodium proton exchanger 3 (NHE3) trafficking

regulation and determine molecular mechanisms of renal proximal tubular sodium

iii

handling, which might contribute to the impaired sodium excretion associated with hypertension and results may help to develop effective therapy for hypertension.

Renal proximal tubules (RPTs) responsible for 65-70% of filtered sodium and water reabsorption have profound effects on renal and body fluid balance associated with hypertension. Studies from our lab were the first to demonstrate that in renal proximal tubular cells, binding of cardiotonic steroids (CTS) such as ouabain to Na/K-ATPase stimulates Na/K-ATPase signaling cascade and induces the redistribution of basolateral

Na/K-ATPase and apical NHE3, leading to a net increase in urinary sodium excretion.

The first manuscript entitled “Ouabain-stimulated trafficking regulation of the

Na/K-ATPase and NHE3 in renal proximal tubule cells” improves our understanding of Na/K-ATPase and NHE3 trafficking regulation. Three renal proximal tubular lines (human HK-2, porcine LLC-PK1, and AAC-19 originated from LLC-PK1 cells in which the pig α1 was replaced by ouabain- resistant rat α1) were employed to compare ouabain-induced regulation of the α1 subunit and NHE3 as well as transcellular 22Na+ transport. Data indicate that our previous observations that ouabain-induced redistribution of Na/K-ATPase α1 subunit and NHE3 is not species-specific. In LLC-PK1 cells, ouabain also inhibited the endocytic recycling of internalized NHE3, but has no significant effect on recycling of endocytosed α1 subunit.

The second manuscript entitled “Impairment of Na/K-ATPase signaling in renal proximal tubule contributes to Dahl salt-sensitive hypertension” defines a

iv

novel mechanism of salt-sensitive hypertension. Studies from Sprague Dawley and

Dahl salt-resistant and Dahl salt-sensitive rats demonstrate that impaired Na/K-

ATPase signaling and consequent regulation of Na/K-ATPase and NHE3 in renal proximal tubule may contribute to salt induced hypertension in the Dahl S rat.

Given there is no difference in the Na/K-ATPase α1 gene (Atp1a1) coding and α1 sensitivity to ouabain between Dahl salt-resistant and Dahl salt-sensitive rats, other factors must be present to affect the activation of Na/K-ATPase/c-Src signaling.

Reactive oxygen species (ROS) play a critical role in the pathogenesis of hypertension. Manuscript 3 entitled “Redox modulation of the Na/K-ATPase signaling and renal proximal tubular sodium handling” present that ouabain- stimulated Na/K-ATPase signaling and subsequent redistribution are redox- sensitive, and ouabain induces direct carbonylation of proline/threonine residues in the actuator (A) domain of the Na/K-ATPase α1 subunit. These data indicate that a proper redox modulation of renal proximal tubular Na/K-ATPase signaling is critical in renal sodium handling and might play an important role in counteracting volume expansion mediated high blood pressure.

All in all, this dissertation not only improves our understanding of Na/K-

ATPase signaling regulation, but also defines a novel mechanism involved in salt- sensitive hypertension, and also demonstrates ouabain-activated Na/K-ATPase signaling is redox sensitive, which might play a crucial role in the regulation of renal sodium handling and blood pressure.

v

Dedication

I dedicate this dissertation to my lovely family, particularly to my understanding and patient husband, Dengyue, who has put up with my so many years of research overseas and has supported my every step of the way, and to our precious daughter Shuyan, who is the joy of our lives, and to my loving parents who have never failed to give me moral support and encouragement.

vi

Acknowledgments

I wish to extend my sincere appreciation to all the persons who made my

dissertation possible. I would never have been able to finish my dissertation without the

guidance of my committee members, help from friends and support from my family.

I would like to express my sincere gratitude to Dr. Joseph Shapiro, my major advisor for giving me a chance to join his excellent lab and providing me a fascinating research topic. I cannot thank him enough for his patience, his solicitude, his kindness, his wisdom and support. His inspiring words and excellent guidance will sway my life deeply and make me achieve what I have dreamt of.

I am very grateful to Dr. Jiang Liu, for his thoughtfulness and generosity with time and energy, for his sharing meticulous research and insights with me that support and expand my thesis work, for risking his health to help me with those critical data.

I am also indebted to my committee members, Dr. Nader Abraham, Dr. Bina Joe,

Dr. Deepak Malhotra, and Dr. Zijian Xie, for serving on my dissertation committee, for their expert suggestions, contributions and good-natured support throughout the production of my research and dissertation.

vii

I would also like to thank my fellow lab members, past and present, for their assistance and support over the years: Dr. Steven Haller, Dr. David J. Kennedy, Dr. Jiang

Tian, Dr. Christopher Drummond, Sandeep Vetteth, Shalini Gupta, Mohammad Taleb,

Vinai Katragadda, Anna Shapiro, Chiamaka Mbaso, Georgy Budnyy, Imad Hariri, Sayed

Moustafa Helmy, Eric Kim. I really enjoy being with all of you in the lab. Special thanks

go to Dr. Sankaridrug Periyasamy, our previous lab manager, and his wife for their support and assistance when I first came to the lab.

Many thanks must also go to Ms. Carol Woods, and Ms. Jenelle Thurn for their research assistance; to Dr. Robert Blumenthal for his timely help with my graduate admission process, to Dr. Randall Ruch for his support and assistance as an assistant dean

for Admissions, Biomedical Graduate Program, to Dr. Andrew Beavis for his course and

research help as a director of CVMD track; to Dr. Lijun Liu, Dr. Jean H. Overmeryer and

Dr. John David Dignam for their kind, unselfish help to make my experiments go well, to

Dr. Nancy Collins, my mentor in Graduate Student Professional Mentoring Program, for

her valuable advice in many different ways, to Madge Levinson, my English tutor, for her

encouragement and guidance with my English language study.

I would like to gratefully acknowledge the support, encouragement and friendship

of my good friends, classmates and co-workers over the years: Lauren Stanoszek, Erin

Crawford, Xiaolu Zhang, Jiyoun Yeo, Jieying Wang, Shuai Dong, Lu, Shuo Geng,

Marjie Gable, Jian Wu, Yan Bai, Daxiang Li, Yiliang Chen, Zhichuan Li, Qiying Ye,

Zhen Jiang, Fangfang Lai, Katye Smedlund, Jean-Yves Tano, Joshua Waldman, Qiang

Mei, Jun Lu, Jie Zhou. viii

Additionally, I wish to thank the members of Dr. Nader Abraham, Dr. Bina Joe, Dr.

Lijun Liu and Dr. Zijian Xie’s lab for sharing their valuable thoughts with me, helping me get through difficult experiment.

I would like to thank my parents, my family and all my friends for their steadfast support and encouragement throughout the years. Lastly, but most importantly, I would like to thank my loving husband, Dengyue Sun, and daughter, Shuyan Sun for never failing to lift my spirits. Your love and support have been with me through all the good

times and bad, and helping to keep me sane.

ix

Contents

Abstract iii

Dedication vi

Acknowledgments vii

Contents x

List of Abbreviations xiii

Chapter 1 – Literature Review 1

Chapter 2 – “Ouabain-Stimulated Trafficking Regulation of the Na/K-ATPase and

NHE3 in Renal Proximal Tubule Cells” (Manuscript 1accepted for publication) 55

2.1 Abstract 56

2.2 Introduction 57

2.3 Experimential Methods 58

2.4 Results 64

2.5 Discussion 68

2.6 Manuscript References 73

2.7 Figure Legends 79 x

2.8 Tables and Figures 82

Chapter 3– “Impairment of Na/K-ATPase Signaling in Renal Proximal Tubule

Contributes to Dahl Salt-Sensitive Hypertension” (Manuscript 2) 90

3.1 Abstract 91

3.2 Introduction 92

3.3 Materials and Methods 92

3.4 Results 97

3.5 Discussion 100

3.6 Manuscript References 105

3.7 Figure Legends 109

3.8 Tables and Figures 112

Chapter 4 – “Redox Modulation of the Na/K-ATPase Signaling and Renal Proximal

Tubular Sodium Handling ” (Manuscript 3 to be submitted) 122

4.1 Abstract 123

4.2 Introduction 124

4.3 Materials and Methods 125

4.4 Results 131

4.5 Discussion 139

4.6 Manuscript References 144

4.7 Figure Legends 152

4.8 Figures 156 xi

Chapter 5 – Summary and Conclusions 174

xii

List of Abbreviations

BP------Blood pressure

CAT------Catalase

C2-9-------1 knock out cells

CO------Carbon oxide

CoPP------Cobalt protoporphyrin

CTS------Cardiotonic steroids

Dahl R rat------Dahl salt-resistant rat

Dahl S rat------Dahl salt-sensitive rat

EGFR------Epithelial growth factor receptor

GPx------Glutathione peroxidase

G.O. ------Glucose oxidase

Grb2 ------Growth factor receptor-bound 2

HO-1------Heme oxygenase 1

HS------High salt

LLC-PK1------Porcine renal proximal tubule cell lines

LS------Low salt

MAPK------Mitogen-activated protein kinase

MBG------Marinobufagenin

MEK------MAPK-ERK activating kinase

NAC------N-acetyl cystein xiii

NHE3------Sodium proton exchange 3

NKA------Na/K-ATPase

NO------Nitric oxide

PKC------Protein kinase C

PLC------

PI3K------Phosphoinositide 3’ kinase

PY-17 cells------NKA knock down cells

RPT------Renal proximal tubules

ROS------Reactive oxygen species

Shc------Src homology collagenlike protein

SNS------Sympathetic nervous system

SOD------Superoxide dismutase

SOS------Son of sevenless

SYF------Mouse fibroblast cells with deficient Src kinase

SYF+c-Src------Mouse fibroblast cells overexpressing Src kinase

xiv

Chapter 1-Literature Review

1. Na/K-ATPase (EC 3.6.3.9, also known as Na pump)

Na/K-ATPase, originally reported by Skou (Skou 1957), is ubiquitous and crucial important membrane protein that maintains the high internal K+ and low internal Na+ concentrations via coupling the transmembrane transport of three Na+ outward and two

K+ inward utilizing energy derived from ATP hydrolysis (Post, Hegyvary et al. 1972;

Lingrel and Kuntzweiler 1994; Blanco and Mercer 1998). In addition to its classical

pumping function, The Na/K-ATPase acts as a signal transducer that relays extracellular

messages into the cell through regulation of protein-protein interactions (Xie 2003;

Schoner and Scheiner-Bobis 2005), which can subsequently up or down-regulate various cellular processes such as gene activation, motility, cell-cell contact, cell proliferation and apoptosis, as well as transcellular transport processes in intestine, glands, and kidney

(Schoner and Scheiner-Bobis 2007; Bagrov and Shapiro 2008; Newman, Yang et al.

2008).

1.1 Na/K-ATPase structure

P-type including Na+, K+-ATPase, Ca2+-ATPas (SERCA) and gastric

H+, K+-ATPase are responsible for the active transport of a variety of cations across the

1 membranes (Sachs and Munson 1991; Lutsenko and Kaplan 1995). P-ATPase is

designated due to catalyzing auto- or self- phosphorylation of a key conserved aspartate

residue within the pump. The Na/K-ATPase, the largest protein complex in the family of

P-type cation pump, is an oligomer consisting of two major polypeptides, α- and β- subunits, and a tissue-specific auxiliary regulatory subunit known as FXYD. A heterodimer of α–subunit and β–subunits is the minimum functional unit of Na/K-

ATPase (Jorgensen and Andersen 1988). In humans, there are four isoforms for the α– subunit (α1-α4), three for the β-subunit (β1-β3) (Lingrel, Orlowski et al. 1990; Axelsen

and Palmgren 1998) and seven for the FXYD (Kuster, Shainskaya et al. 2000; Sweadner

and Rael 2000), leading to different Na/K-ATPase variants.

1.1.1 α subunit

The α subunit is the catalytic unit with a molecular mass of 112 KDa, which

consists of ten transmembrane ((TM)) helices (Figure 1-1, M1-M10) (Blanco and Mercer

1998), three cytoplasmic conserved domains, actuator (A), binding (N), and

phosphorylation (P) (Figure 1-2). ATP binds to the N domain and phosphrylates P

domain. The A domain is involved in this reaction to manipulate the TM segments to

open the occlusion for sodium or potassium binding, depending on which state, E1 or E2,

the protein is in. The lysine-rich N-terminal extension of around 40 residues

phosphorylated by protein kinase C might provide a platform for interacting with other

such as IP3 receptor (Figure 1-2B). The fascinating observation by Toyoshima

team (Toyoshima, Kanai et al. 2011) (Xie and Cai 2003)is that the N-domain is less

(~20°) inclined toward the A-domain, and a salt bridge between the positively charged

residue Arg551 in the N-domain and the negatively charged residue Glu223 in the A-

2 domain is one crucial link between them (Figure 1-2A). Since Arg551 interacts with the

β- of ATP, the salt bridge will be broken by ATP/ADP binding, which speed

up the opening of α subunit headpiece and then transition into E1 (Shinoda, Ogawa et al.

2009).

Figure 1-1: Scheme of the membrane topology of the α and β isoforms of the Na/ K-ATPase (Blanco and Mercer 1998). Sequences of rat α and β isoforms are shown. Residues are colored to indicate the amino acid homology among the different α isoform or β isoforms.

In addition, the α-subunit contains the binding sites for the cations, ATP and the inhibitors such as ouabain (Pedemonte and Kaplan 1990; Mercer 1993; Lingrel and

Kuntzweiler 1994; Pressley 1996). The binding sites for Na+ and cardiotonic steroids

(CTS) are on the extracellular segments and the binding sites for K+ and ATP are on the

intracellular loops. Specifically, the biding sites for the inhibitors CTS include TM1-TM2,

3 TM5-TM6, and TM7-TM8 loops and several amino acids from the transmembane regions M4, M6, and M10. The biding site for ATP is located in the pocket formed by the intracellular loop TM4-TM5. The proximal and distal regions of intracellular loop TM4-

TM5 are the phosphorylation domains. Besides, of the ten TM helices, M4-M6 and M8 contain the binding sites for the cations. The M7-M10 helices have special function and might undergo large-scale rearrangement in Na/K-ATPase. The cytoplasmic half of M7 is distinctly kinked in Na/K-ATPase. The outside of M9 interacts with FXYD, whereas

M10 associates with TM helix of the β subunit (Figure 1-2) (Bagrov, Shapiro et al. 2009;

Toyoshima, Kanai et al. 2011).

1.1.2 β subunit

The β subunit is a heavily glycosylated polypeptide that crosses the membrane once (Blanco and Mercer 1998) and has a molecular weight between 40 and 60 KDa depending on the degree of glycosylation in different tissues. The β subunit is composed of a short N-terminal cytoplasmic domain, one transmembrane helix located in the groove between the M7 and M10 helices of the α-subunit and a large extracellular domain that almost covers the entire extracellular surface of the α-subunit (Figure 1-1.

Figure 1-2A).

The primary role of the β subunit is to function as a chaperone, facilitating routing of the α-subunit to the plasma membrane and stabilizing its conformation (Chow and

Forte 1995; Geering 2001; Geering 2008). In addition, the β subunit is also involved in the occlusion of K+ and the modulation of the K+ and Na+ affinity of the (Jaisser,

4 Horisberger et al. 1992; Lutsenko and Kaplan 1993; Blanco, Koster et al. 1995; Eakle,

Lyu et al. 1995; Geering 2001).

1.1.3 FXYD (γ subunit)

The FXYD protein, composing of one transmembrane helix, one N-terminal

extracellular region and a cytoplasmic part, is a small polypeptide of 8-14KDa (Blanco

and Mercer 1998). γ subunit is termed for FXYD in kidney outer medullar (Morth,

Pedersen et al. 2007). It interacts with the αβ-complex as a third subunit and modulates

the affinities of Na+, K+ and ATP, and then regulates Na/K-ATPase in a tissue- and

isoform-specific manner (Blanco and Mercer 1998; Cornelius and Mahmmoud 2003;

Geering 2006). However, it is unclear about how various FXYD proteins affect binding

of Na+/K+, and the kinetic properties of Na/K-ATPase (Blanco and Mercer 1998;

Toyoshima, Kanai et al. 2011).

1.2 Na/K-ATPase function (Post-Albers Model)

The Na/K-ATPase belongs to P-ATPases (known as E1-E2 ATPases) that is composed of two main conformations called E1 and E2, which are responsible for the

active transport of a variety of cations across cell membranes. In the E1 state, the TM cation binding sites have high affinity for Na+ and face the cytoplasm. In E2, the binding

sites have low affinity for Na+ but high affinity for K+ and face the extracellular side. In

E1 state, three sodium bind to the pump from the cytoplasmic side of the membrane.

ATP phosphorylates the enzyme (as indicated by the P attached to the cytoplasmic side of the pump), and the remaining ADP is released. The conformation then changes to E2, and the three sodium ions are released into the extracellular solution.

5

Figure 1-2: Architecture of Na/K-ATPase from Shark Rectal Gland with Bound 2− + + MgF4 and K , a Stable Analog of the E2·Pi·2K State (Toyoshima, Kanai et al. 2011). (A) A ribbon diagram of Na/K-ATPase with ouabain (shown in space fill) bound at low affinity (PDB ID: 3A3Y). Color changes gradually from the N-terminal (blue) to the C- terminal (red). ATP is taken from the E2(TG)·ATP crystal structure of Ca2+-ATPase (SERCA1a) (PDB ID: 3AR4) and docked in the corresponding position. Bound K+ ions are marked (I, II, and C) and circled. Inset shows a simplified diagram of the Post-Albers scheme. CLR, cholesterol; OBN, ouabain. (B) Digestion sites (proteinase K, black “x”; chymotrypsin, magenta “x”) and interaction sites with other proteins (dotted circles). Residue numbers in parentheses refer to corresponding residues in SERCA1a. AP-2, adaptor protein 2; IP3R, inositol 1,4,5-triphosphate receptor; PI3K, phophoinositide-3 kinase.(C) Disposition of transmembrane helices viewed approximately perpendicular to the membrane from the cytoplasmic side (Na/K-ATPase, yellow; SERCA1a, cyan). CLR, cholesterol.

After two potassium ions bind to the pump from the extracellular side, the pump is dephosphorylated and returns to E1 state. ATP binding occurs and the potassium is released into the intracellular solution (Figure 1-2, inset) (Albers 1967; Post, Hegyvary et al. 1972).

6 2. Na/K-ATPase ligand – Cardiotonic steroids

Cardiotonic steroids (CTS), also referred to as digitalis-like factors, are inhibitors of

Na/K-ATPase. CTS are composed of plant-derived ouabain, digoxin, and vertebrate- derived marinobufagenin (MBG). First and perhaps foremost, many endogenous CTS have been purified and characterized in animals and human (Hamlyn, Blaustein et al.

1991; Lichtstein, Gati et al. 1993; Bagrov and Fedorova 1998; Komiyama, Dong et al.

2005). Both endogenous ouabain and MBG synthesized in and released from hypothalamus and adrenal contex could be regulated by ACTH, angiotensin, vasopressin, and phenylephrine (Laredo, Shah et al. 1997). Generally, endogenous CTS circulate in a protein-bound form with plasma levels from subnanomolar to nanomolar concentration in different condition (Bagrov, Shapiro et al. 2009).

The α subunit of Na/K-ATPaseh as a specific for CTS on the extracellular loops (TM1-TM2, TM5-TM6, and TM7-TM8). Extracellular TM1-TM2 loop (amino acids from 111-122 sequences) form the most important part of the putative

CTS binding site.

The sodium pump has a different sensitivity to CTS due to the different amino acid sequence. CTS, on the other hand, display different affinity to different isoforms. For instance, a nanomolar concentration of bufadienolide marinobufagenin can inhibit Na/K-

ATPase in rodent renal epithelial cells consisting of nearly exclusively the α1 isoform, whereas ouabain is much less active with renal Na/K-ATPase (Bagrov, Shapiro et al.

2009).

3. Na/K-ATPase signaling

7 Na/K-ATPase has three classic features that are the pump, the enzyme, and the receptor to CTS. Quite importantly, low concentrations of CTS, for instance, circulating

levels from subnanomolar to nanomolar in humans that do not inhibit the enzymatic

function of sodium pump, are able to initiate its signaling pathway (Liu, Periyasamy et al.

2002), which plays a key role in genomic and nongenomic functions. Moreover, the

following studies provide the positive evidence supporting the signaling function for the

Na/K-ATPase. First, no change in cytosolic [Na+] is measured in mammalian cells

exposed to those concentrations of CTS (Liu, Tian et al. 2000). Second, the caveolar

Na/K-ATPase is closely related to the key signaling proteins such as c-Src and EGFR

instead of pumping sodium (Liang, Cai et al. 2006). Third, the phosphorylation of key

signaling proteins caused by CTS can also be observed in cell-free systems, in which no

[Na+] is changed (Wang, Haas et al. 2004). Although increasing evidence demonstrates

that Na/K-ATPase signaling effects might be independent of its inhibition of transmembrane sodium transport (Xie and Askari 2002; Schoner and Scheiner-Bobis

2007; Tian and Xie 2008), we propose that the classic and signaling pathway may work both in parallel and synergistically to affect physiological consequence of Na/K-ATPase

(Bagrov, Agalakova et al. 2009).

The Na/K-ATPase signaling pathway involves the association of c-Src with the

Na/K-ATPase in a caveolar domain. Binding of CTS to the Na/K-ATPase stimulates c-

Src activation, which, in turn, transactivates the EGFR and phospholipase C (PLC), leading to a sequential cascade that involves generation of reactive oxygen species (ROS), activation of mitogen-activated protein kinase (MAPK) through activation of its mitogen- activated protein kinase kinase (MEK), activation of PI3K, and activation of protein

8 kinase C (PKC) (Figure 1-3). The activated pathways that exist in different cells including

renal proximal tubular cells, cardiac myocytes and whole animal models, will generate the

genomic and nongenomic effects (Xie and Cai 2003; Bagrov, Shapiro et al. 2009).

Figure 1-3: Schematic presentation of Na/K-ATPase signalosome (Xie and Cai 2003). The Na+/K+-ATPase (pump) is preassembled with its partners in caveolae; ouabain binding to the pump activates the signalosome, and transduces signals via multiple pathways. EGFR, epithelial growth factor receptor; PKC, protein kinase C; PI3K, phosphoinositide 3’ kinase; Grb2, growth factor receptor-bound protein 2; SOS, son of sevenless; Shc, src homology collagenlike protein; PLC, phospholipase C; MAPK, mitogen-activated protein kinase; MEK, MAPK-ERK activating kinase; ROS, reactive oxygen species.

It is noteworthy that Src activation is the critical step for Na/K-ATPase signaling.

Under basal circumstance, the caveolar Na/K-ATPase binds to Src and maintains Src

inactivated. The binding of Na/K-ATPase to CTS will cause an alternation of

conformation, which allows the Src to be activated (Figure 1-4) (Tian, Cai et al. 2006).

4. Na/K-ATPase signaling and renal sodium handling

4.1 Sodium proton exchanger 3 (NHE3)

9

Figure 1-4: Schematic presentation shows how ouabain regulates the Na/K- ATPase/Src receptor complex (Tian, Cai et al. 2006).

There are nine NHE isoforms in the mammalian genome. NHE 1-5 reside predominantly in the plasma membrane and NHE 6-9 are expressed largely in

endomembrane organelles (Alexander and Grinstein 2009; Donowitz, Mohan et al. 2009).

They have various tissue distribution and function. In the renal proximal tubules, NHE3

(SLC9A3) presented along apical/luminal brush border membrane is responsible for two-

third of filtered sodium and fluid reabsorption as well as maintenance and regulation of intravascular volume and BP (Biemesderfer, Pizzonia et al. 1993; Amemiya, Loffing et al.

1995; Alexander and Grinstein 2006). It is widely accepted that apical NHE3 is the rate- limiting step of apical Na+ entry and sodium reabsorption. In other words, NHE3 is a

major Na+ uptake mechanism in the proximal tubules, and increased activity of NHE3 in the renal brush border membrane is involved in the pathogenesis of hypertension

(Morduchowicz, Sheikh-Hamad et al. 1989). Conversely, NHE3-deficient mice are hypotensive and develop acidosis (Schultheis, Clarke et al. 1998; Lorenz, Schultheis et al.

1999; Ledoussal, Lorenz et al. 2001) because of reduced Na+ reabsorption and increased

Na+ excretion in the proximal tubule (Liu and Xie 2010).

10 Rats on low salt exhibit a higher NHE activity compared with rats on high salt diet,

which may contribute to decreased NaCl reabosorption in extracellular fluid volume

expansion, enhanced NaCl reabosorption in extracellular fluid volume depletion (Moe,

Tejedor et al. 1991; Fisher, Lee et al. 2001; Yang, Sandberg et al. 2008). The possible

molecular mechanisms underlying the decreased NHE3 activity in response to high salt is

attributed to the redistribution of NHE3 (Liu, Yan et al. ; McDonough ; Oweis, Wu et al.

2006; Yang, Sandberg et al. 2008). In this scenario, NHE3 plays a protective role

counterbalancing blood pressure rising. For instance, the NHE3 activity decreases with

age in SHR rats (Crajoinas, Lessa et al. ; Yang, Leong et al. 2007).

NHE3 activity is regulated by many factors such as NHE regulatory factor-1

(NHERF-1), angiotension II, sympathetic nervous system, thyroid hormone, insulin and

other humoral agents (Wang, Armando et al. 2009). Importantly, the total number of

NHE3 in the apical membrane and its activity are involved in acute regulation of its

function. The surface expression of NHE3 is mainly regulated by alterations in

endocytosis/exocytosis, and is considered to be the primary regulatory mechanism of

NHE3 activity. NHE3 traffics between the plasma membrane and early/recycling endosomes via a clathrin- and PI3K-dependent pathway (D'Souza, Garcia-Cabado et al.

1998; Kurashima, Szabo et al. 1998; Chow, Khurana et al. 1999; Janecki, Janecki et al.

2000). The NHE3 activity can be stimulated by exocytosis (Collazo, Fan et al. 2000;

Yang, Amemiya et al. 2000; Hu, Fan et al. 2001) and inhibited by endocytosis. Moreover,

activation of Src, PKA, PKC and an increase in intracellular Ca2+ are involved in the

regulation of NHE3 trafficking (Liu and Xie 2010).

11 Our lab has reported that CTS can induce the endocytosis of basolateral Na/K-

ATPase and apical NHE3 coordinately in the renal proximal tubules through Na/K-

ATPase signaling pathway, leading to a decrease in transepithelial sodium reabsorption

(Figure 1-5) (Oweis, Wu et al. 2006; Liu and Shapiro 2007; Cai, Wu et al. 2008). This mechanism contributes to renal proximal tubular sodium handling in response to volume expansion and high blood pressure (see below for details).

4.2 CTS as a 3rd factor and Na/K-ATPase signaling

The “3rd factor”, first described by De Wardener (De Wardener, Mills et al. 1961), is called because it was a factor in addition to aldosterone and glomerular filtration rate

(GFR) in determining renal sodium handling. Bricker postulated that this 3rd factor might be an inhibitor of the Na/K-ATPase (Bricker 1967). Buckalew, et al demonstrated inhibitor of Na/K-ATPase (Buckalew, Martinez et al. 1970) in plasma and plasma ultrafiltrate of volume expanded dogs . Yates et al reported that either ouabain or plasma volume expansion produces modest increases in sodium excretion and urine flow, whereas ouabain combined with plasma volume expansion increases sodium excretion in a synergistic rather than additive manner (Yates and McDougall 1995). Interestingly, high salt diet stimulates an increase in plasma ouabain/MBG and urinary ouabain/MBG excretion (Manunta, Hamilton et al. 2006). In addition, mounting evidence favors the view that a 3rd factor, which is independent of both glomerular filtration rate and aldosterone levels, modulates sodium excretion (Bricker 1967).

Our lab has been focusing on CTS and renal sodium handling for more than ten years. The following in vitro and in vivo studies from our lab suggest that CTS may function as a natriuretic factor through Na/K-ATPase signaling (Liu and Shapiro 2007).

12 (1) Administration of CTS induces a time- and dose-dependent endocytosis of Na/K-

ATPase in LLC-PK1 cells (porcine renal proximal tubular cells) (Liu, Periyasamy et al.

2002). Ouabain, digoxin and MBG have a different efficacy. Specifically, digoxin seems to be the least effective, whereas the others are more effective. (2) CTS inhibit transcellular 22Na+ transport in LLC-PK1 cells. The endocytosis of Na/K-ATPase is

mediated through clathrin-coated pits and activation of Na/K-ATPase signaling cascades

containing Na/ K-ATPase/Src/EGFR (Liu, Kesiry et al. 2004; Liu, Liang et al. 2005). (3)

In vivo studies provide evidence showing CTS-induced endocytosis involving decreased

sodium reabsorption in RPTs after salt loading (Periyasamy, Liu et al. 2005). (4) CTS can

mediate decreases in the apical cell surface NHE3 due to its endocytosis (Oweis, Wu et al.

2006; Cai, Wu et al. 2008).

Much interestingly, several fascinating reports further confirm this concept -CTS

as a natriuretic factor through Na/K-ATPase signaling. Genetically engineered mice with

ouabain-sensitive α1 subunit of Na/K-ATPase display greater natriuretic responses to salt

loading than mice with ouabain-resistant α1 subunit of Na/K-ATPase, confirming that α1

subunit of Na/K-ATPase as ouabain receptor participates in natriuretic response to salt

loading (Dostanic-Larson, Van Huysse et al. 2005; Loreaux, Kaul et al. 2008). In addition,

Nesher and Dvela et al, for the first time, show that endogenous circulatory ouabain

originating in the adrenal has physiological roles controlling vasculature tone and sodium

homeostasis in normal male Wistar rats (Nesher, Dvela et al. 2009). Likewise, another

endogenous MBG has similar natriuretic and vasoconstrictor function (Fedorova,

Kolodkin et al. 2001; Fedorova, Talan et al. 2002; Bagrov and Shapiro 2008).

13

Figure 1-5: Schematic illustration of the effect of CTS-mediated Na/K-ATPase signaling on renal proximal tubular sodium reabsorption (Liu and Xie 2010). NKA, Na+, K+-ATPase.

In summary, studies from our laboratory has demonstrated CTS-mediated redistribution of Na/K-ATPase and NHE3 via activation of Na/K-ATPase signaling cascade might be one of mechanisms linking CTS to a natriuretic factor. Specifically, binding of CTS to Na/K-ATPase stimulate Na/K-ATPase-Src signaling pathway and induce the redistribution of basalateral α1 subunit of Na/K-ATPase and apical NHE3, which mediates sodium and water reabsorption in renal proximal tubules. The downregulation of NHE3 will decrease transepithelial sodium transportation from apical membrane into basalateral membrane, leading to a net increase in urinary sodium excretion (Figure 1-5) (Liu and Shapiro 2007; Liu and Xie 2010).

5. Na/K-ATPase signaling and blood pressure regulation

Accumulating evidence suggests that CTS are important modulators of blood pressure through Na/K-ATPase. Approximately 50% of uncomplicated essential

14 hypertensive patients have an increase in plasma ouabain level (Rossi, Manunta et al.

1995; Manunta, Stella et al. 1999; McKinnon, Lord et al. 2003). Interestingly, some

hypertensive patients with elevated circulating levels of endogenous ouabain do not

appear to be related to plasma renin activity, and may not involve adrenal type-2

dopaminergic receptors (Rossi, Manunta et al. 1995). On the one hand, elevated

endogenous ouabain would lead to vasoconstriction and high blood pressure by inhibition

α2 subunit of Na/K-ATPase, followed by Ca2+ entry via the Na+/Ca2+ exchanger in

vascular smooth muscle cells (Bagrov, Fedorova et al. 1996; Fedorova, Doris et al. 1998;

Manunta, Hamilton et al. 2006; Krzesinski and Cohen 2007; Bagrov and Shapiro 2008;

Bagrov, Shapiro et al. 2009). In kidney, on the other hand, elevated endogenous ouabain

functions as a natriuretic substance by binding to α1 subunit of Na/K-ATPase, followed

by endocytosis of α1 subunit of Na/K-ATPase and NHE3, leading to an increase in urinary sodium excretion (see 4.2 for detail). The latter could counterbalance the

conditions of volume expansion and salt loading, which is consistent with Arthur

Guyton’s following quantitative mathematical explanation.

Based on Arthur Guyton’s quantitative mathematical model for the relation

between blood pressure an natriuresis (pressure-natriuresis relationship) (Guyton 1961;

Guyton and Coleman 1999), salt loading would transiently increase blood pressure,

which in turn would raise sodium excretion until the baseline steady state pressure is

reached. When the blood pressure/nariuresis relationship is shifted to the right, higher

blood pressure values are required to enable the kidney to excrete sodium loads (Figure

1-6).

15

Figure 1-6: Different renal set points for natriuresis in normotensive and hypertensive patients. When the curve shifts to the right in impaired pressure natriuresis, the equilibrium point shifts and the mean arterial pressure is elevated.

Taken together, Na/K-ATPase appears to have a significant effect on the relationship between blood pressure and natriuresis.

When it comes to the relationship between ouabain and MBG in the regulation of blood pressure, our co-laboratory postulated that brain endogenous ouabain caused by salt loading activates the central renin-angiotensin system (RAS) in the hypothalamus and pituitary, which in turn stimulates the RAS in the adrenal cortex via sympathetic nervous system (SNS) activation. Activation of angiotensin II facilitates the production

and release of MBG in adrenal cortex with a primary adaptive aim to induce natriuresis

through inhibition of renotubular Na/K-ATPase. Excessive MBG production, however,

induces vasoconstriction by inhibition of α2 subunit of Na/K-ATPase in vascular smooth

muscle cells (Fedorova, Agalakova et al. 2005; Bagrov, Shapiro et al. 2009). The above

postulation is supported by the following observations: (1) a transient increase in

circulating endogenous ouabain precedes a sustained increase in circulating MBG in Dahl

salt-sensitive rats after both acute and chronic salt loading (Fedorova, Lakatta et al. 2000;

16 Fedorova, Talan et al. 2002); (2) peak transient responses is observed in the amygdala,

hippocampus, and supraoptic nucleus of hypothalamus in response to salt loading in Dahl

salt-sensitive rats (Fedorova, Zhuravin et al. 2007); (3) intrahippocampal microinjection of low dose of ouabain (30pg in 0.5µl in each hemisphere) mimicked the above events completely (Fedorova, Zhuravin et al. 2007); (4) normotensive humans have a transient increase in urinary endogenous ouabain after salt loading, followed by a more sustained

increase in renal marinobufagenin excretion (Anderson, Fedorova et al. 2008; Bagrov and

Shapiro 2008).

6. Reactive oxygen species, Na/K-ATPase signaling and renal sodium handling

6.1 Reactive oxygen species (ROS) and Na/K-ATPase

- ROS, including superoxide anions (O2 ), hydroxyl radicals (·OH) and hydrogen

peroxide (H2O2), are generated as a natural byproduct of normal metabolism from

mitochondria and peroxisomes, as well as from various cytosolic enzyme systems (i.e. xanthine-xanthine oxidase and NADPH oxidase). Antioxidant systems that remove ROS

− encompasse superoxide dismutases (SOD), which converts O2 to H2O2, and catalase that

convert to water, as well as glutathione peroxidase (GPx), which can reduce lipid

hydroperoxides to their corresponding alcohols and to reduce to water. Besides, free radical scavengers such as vitamin C and E also limit ROS. In a word, an antioxidant system can regulate overall ROS levels to maintain physiological homeostasis (Halliwell and Gutteridge 1990).

- Particularly, ·O2 and H2O2 appear to be more important in cardiovascular cells

- (Paravicini and Touyz 2008). ·O2 is short-lived. It can be produced by the reduction of

17 molecular oxygen by NADPH oxidase. Further reduction of superoxide, by superoxide

dismutase, results in the formation of H2O2 (Curtin, Donovan et al. 2002; England,

O'Driscoll et al. 2004). Therefore, the main source of H2O2 in vascular tissue is the

- - dismutation of ·O2 either spontaneously or by SOD. ·O2 is unable to cross cellular

membranes owing to its charge, except possibly through channels. In contrast, H2O2

- has a longer biological lifespan than ·O2 . It is relatively stable and is easily diffusible

within and between cells (Paravicini and Touyz 2008).

It is worth to note that ROS can induce a number of nonenzymatic modifications of proteins such as carbonylation, nitrotyrosin, o-tyrosine, and dityrosine. Protein

carbonylation is widely used a marker for oxidative stress (Chevion, Berenshtein et al.

2000). ROS cause the oxidation of amino acid side chains on proteins, leading to exact

conformation and pattern of folding transition, which are tightly connected to their activity and function (Berlett and Stadtman 1997; Chevion, Berenshtein et al. 2000;

Stadtman and Levine 2000; Wondrak, Cervantes-Laurean et al. 2000).

Although excessive generation of ROS is related to cell injury in a variety of pathological conditions including those of the cardiovascular system, mounting evidence indicates that ROS also serve as second messengers within several signal pathways involved in the control of gene (Sen and Packer 1996; Lander 1997). To our interest, an increase in ROS by hypoxia can induce the endocytosis of Na/K-ATPase through reducing Na/K-ATPase activity at the cell surface (Dada and Sznajder 2003).

In a word, an increase in ROS levels may also activate diverse signaling pathways, which may have either damaging or potentially protective functions (Miller, Suzuki et al. ;

Finkel and Holbrook 2000).

18 6.1.1 Na/K-ATPase sensitivity to ROS

Na/K-ATPase is one of the targets for ROS (Lees 1991; Boldyrev, Bulygina et al.

1995). ROS interact with Na/K-ATPase and induce its conformational change, which is

associated with oxidant sensitivity (Xie, Wang et al. 1990; Huang, Wang et al. 1994).

The sensitivity of Na/K-ATPase is dependent on its different isoforms in various tissues

(Xie, Jack-Hays et al. 1995). The α2 and α3 subunit of Na/K-ATPase appear to be more sensitive to H2O2 than the α1 subunit. More sensitive to oxidants the Na/K-ATPase

isoform is, more susceptible to degradation it is (Xie, Jack-Hays et al. 1995). In addition,

brain Na/K-ATPase has more sensitivity than kidney Na/K-ATPase to inhibition by H2O2

(Kurella, Tyulina et al. 1999), while similar sensitivity to oxidants has been found

between ouabain-sensitive α1 (canine) and insensitive α1 (rat), indicating that the

sensitivity of Na/K-ATPase isoform seems not to be species specific, but to be tissue-

dependent.

Taken together, oxidant sensitivity of Na/K-ATPase is related to structural features,

rather than to features that control ouabain sensitivity. Different sensitivity of the Na/K-

ATPase isoform to oxidants appear to depend on the primary sequences of different

subunits and different compositions of subunits in various tissues (Huang, Wang et al.

1994).

6.1.2 Oxidative modification of Na/K-ATPase and its functions

Na/K-ATPase is redox-sensitive (redox, short for reduction/oxidation). H2O2

induces oxidation of Na/K-ATPase, which could be prevented by antioxidants including

the hydrophilic natural antioxidant carnosine, the hydrophobic natural antioxidant alpha-

tocopherol, and the synthetic antioxidant ionol as well as the ferrous ion chelating agent

19 deferoxamine (Kurella, Tyulina et al. 1999). Besides, studies in cerebellar granule cells

(Petrushanko, Bogdanov et al. 2006) suggest that redox state is a potent regulator of the

Na/K-ATPase function. (1) Transport activity of the enzyme is maximal within in a narrow range of redox potentials. Shifts from an "optimal redox potential range" to higher or lower levels cause suppression of the sodium pump activity. (2) Oxidized Na/K-

ATPase (α/β) and FXYD proteins induced by ROS not only inhibit the Na/K-ATPase activity, but also promote its susceptibility to degradation by proteasomal and endosomal/lysosomal proteolytic pathways in different types of cells containing cardiac myocytes, vascular smooth muscle cells, and renal proximal tubular cells (Huang, Wang et al. 1992; Zolotarjova, Ho et al. 1994; Thevenod and Friedmann 1999).

Recently, Rasmussen and co-workers demonstrated that redox signaling mediated by angiotensin receptors and β adrenergic receptors induces oxidative modification

(glutathionylation) of cysteine residue in the β1 subunit of Na/K-ATPase, which could be a physiologic means of regulating protein function. Interestingly, oxidative modification is reversible as phosphorylation does (Figtree, Liu et al. 2009; Rasmussen, Hamilton et al.

2010). However, some studies show that purified enzyme has also been shown to be irreversibly inhibited in response to hydrogen peroxide, the superoxide anion, and the hydroxyl radical (Huang, Wang et al. 1992).

6.1.3 Oxidative modification of Na/K-ATPase and Na/K-ATPase structural regulation

In dog kidney, oxidative modification of Na/K-ATPase by H2O2 induces a decrease in the amount of sulfhydryl (SH) groups, followed by oligomeric structure formation of

Na/K-ATPase, resulting in 50% inhibition of its activity (Boldyrev and Kurella 1996;

20 Dobrota, Matejovicova et al. 1999). Furthermore, the oxidative stability of Na/K-ATPase

is depend on the differences in the number, location, and accessibility of SH groups in

Na/K-ATPase isozymes (Kurella, Tyulina et al. 1999). Besides, the cysteine residues

located in α subunit cytosolic loops is associated with its redox sensitivity.

In the future, studies will be investigated to help characterize changes in the Na/K-

ATPase structure and its possible chemical modification that occurs in response to redox stress.

6.2 ROS involved in Na/K-ATPase signaling

Binding of CTS to the Na/K-ATPase causes cellular ROS production and its downstream effects according to the following observations (Liu, Kennedy et al. 2012).

(1) Ouabain, an inhibitor of Na/K-ATPase, stimulates ROS generation in isolated rat myocytes, pancreatic islets and other cells (Liu, Tian et al. 2000; Kajikawa, Fujimoto et al. 2002; Boldyrev, Bulygina et al. 2003; Huang, Chueh et al. 2004). Moreover, infusion of CTS (MBG) induces ROS generation in experimental animals (Kennedy, Vetteth et al.

2006). (2) ROS induced by ouabain are essential second messengers in ouabain-initiated pathway (Xie, Kometiani et al. 1999). (3) In Na/K-ATPase signaling cascade (Figure 1-

2+ 3), Ras-Raf-MEK-ERK activation increases [Ca ]i, resulting in opening of mitochondrial

ATP-sensitive K+ channels (Tian, Liu et al. 2003) and generation of mitochondrial ROS

(Xie, Kometiani et al. 1999; Liu, Tian et al. 2000). Conversely, ROS stimulate the

signaling function of Na/K-ATPase, followed by a positive amplification loop due to the

mitochondrial production of ROS caused by activation of signaling function of Na/K-

ATPase (Xie, Jack-Hays et al. 1995; Xie and Cai 2003).

21 There is growing convincing data showing the key role of Na/K-ATPase signaling in regulating ROS production. (1) c-Src as a critical initiating step of Na/K-ATPase signaling, is also redox-sensitive and required for ROS generation (derived from NADPH oxidases) (Seshiah, Weber et al. 2002; Touyz, Yao et al. 2003). (2) Lipid rafts are necessary in Na/K-ATPase signaling. In coronary endothelial cells, clustering of lipid rafts on the membrane promotes aggregation and activation of NADPH oxidase, forming a platform for redox signaling through ROS (Touyz 2006; Zhang, Yi et al. 2006). (3) The

Na/K-ATPase regulates caveolin-1 endocytic trafficking and stabilizes the caveolin-1 plasma membrane pool (Cai, Wu et al. 2008). Caveolin 1 is also required in ROS- dependent Angiotensin II signaling (Zuo, Ushio-Fukai et al. 2005).

6.3 ROS in regulation of renal sodium handling and blood pressure

Oxidative stress has been shown to regulate blood pressure and renal sodium handling in various animal models. Physiologically, ROS are necessary in normal redox signaling, while pathological levels of ROS are responsible for renal and vascular dysfunction and remodeling due to oxidative damage (Touyz 2004; Wilcox and

Gutterman 2005; Paravicini and Touyz 2006). Genetic factor(s) partially contribute to high basal ROS levels and the development of hypertension (Lacy, O'Connor et al. 1998;

Lacy, Kailasam et al. 2000).

As mentioned above, CTS-stimulated Na/K-ATPase signaling contributes to RPT sodium handling. ROS serve as important intracellular and extracellular second messengers to regulate many signaling molecules containing Na/K-ATPase signaling.

22 Here we review the possible role of ROS in renal sodium handling and blood pressure regulation.

6.3.1 ROS and renal salt handling

6.3.1.1 ROS and renal NADPH oxidases

A major source of ROS in vascular tissues and kidney is NADPH oxidase (Nox).

Nox originally found in , is an enzymatic complex consisting of seven subunits: membrane subunits p22phox and gp91phox (also termed “Nox2”) (Babior 2004),

cytosolic components p40phox, p47phox, and p67phox, and a low-molecular-weight

Rac1 or Rac2 (“phox” stands for phagocyte oxidase) (Burritt, Quinn et al. 1995;

Fontayne, Dang et al. 2002; Vignais 2002; Paravicini and Touyz 2008). Generally, in

unstimulated cells, p22phox and gp91phox occur as a heterodimeric flavoprotein, cyto- chrome b558. On stimulation, p47phox becomes phosphorylated and the cytosolic

subsunits form a complex that translocates to the membrane, where it associates with

cyto-chrome b558 to assemble active oxidase, transferring electrons from the substrate to

O2, forming superoxide (Touyz, Yao et al. 2003; Paravicini and Touyz 2008). This

process is also regulated by Rac2 and Rac1(Bokoch and Zhao 2006).

Seven members in the Nox family include Nox1, Nox2, Nox3, Nox4, Nox5, Duox1,

and Duox2 (Griendling 2006). They are expressed in all tissues and mediate a variety of

biological functions. Of the Nox family, Nox 4 (originally termed Renox due to its high

− expression in kidney tissue) has been shown to act as an O2 producing NADPH oxidase in vascular smooth muscle and kidney (Geiszt, Kopp et al. 2000; Touyz, Chen et al.

2002). Interestingly, Nox4 requires p22phox for activity, but does not require the assembly

23 of cytosolic subunits (Rivera, Sobey et al. 2010). Nox4 might be located in a number of

cellular components, containing the mitochondria (Graham, Kulawiec et al. 2010),

endoplasmic reticulum in addition to the plasma membrane (Yang, Lane et al. 2009;

Zhang, Zhang et al. 2011). Nox4 and p22phox co-localize to internal membranes where

superoxide generation occurs. This Nox4 and /or p22phox complex would constitute

significant levels of hydrogen peroxide involved in intra- and intercellular signaling

pathways affecting basic cellular functions (Martyn, Frederick et al. 2006). That is why

Nox4 does not require the assembly of cytosolic subunits. In the macula densa cells,

Nox2 is primarily responsible for NaCl-induced superoxide production, whereas Nox4 is major source of basal superoxide (Zhang, Harding et al. 2009).

6.3.1.2 ROS and renal proximal tubular sodium transport

Renal Na+ excretion must be tightly regulated to maintain body fluid balance. In

kidney, the amount of Na+ filtered is several times higher than Na+ intake. More than

99% of filtered Na+ must be reabsorbed and approximately 60–70% of this reabsorption occurs in the proximal tubules. Importantly, the NADPH oxidases are the main resource of ROS and present in the renal cortex, medulla, and vasculature (Shiose, Kuroda et al.

2001). Especially, NADPH oxidases are highly expressed in renal proximal tubules.

(Geiszt, Kopp et al. 2000) and play a critical role in sodium reabsorption.

In general, the Nox family NADPH oxidases transfer electrons from

− NADPH/NADH to oxygen across biological membranes, forming superoxide. O2 can then react with both enzymatic and nonenzymatic cellular constituents to form other

− radicals with biological activity such as H2O2 or OH , or with NO to produce

24 OONO−(Traylor and Mayeux 1997). As shown in Fig. 1-7 (Schreck and O'Connor 2011),

− + superoxide (O2· ) generated by NAD(P)H oxidase inhibits Na uptake into the proximal

tubular cell through inhibiting NHE3 (Na+/H+) and Na+-glucose cotransporters

+ − + + (Na :Glucose). The actions of O2· on other apical Na transporters including Na -

ammino acid coransporters (Na+:AA) and Na+-phospate cotransporters (Na+:Pi) remains

− unknown. O2· reacts with nitric oxide (NO) to produce peroxynitrite (OONO·−), which

inhibits basolateral Na/K-ATPase (Schreck and O'Connor ; Zhang, Imam et al. 2002;

Schreck and O'Connor 2011).

Figure 1-7: Diagramatic sketch indicating the actions of NAD(P)H oxidase on sodium transport in a proximal tubule cell (Schreck and O'Connor 2011). Black arrows represent direction of action/reaction only; thin dashed arrows indicate direction of Na+ transport; red arrows indicate an inhibitory action. Panico et al have also demonstrated that superoxide produced by NADPH oxidase

inhibits proximal tubular fluid reabsorption through reduced NHE3 activity in

spontaneously hypertensive rat (SHR) (Panico, Luo et al. 2009). But inhibition of

NADPH oxidase does not have any effect on fluid absorption in renal proximal tubules

25 from normotensive WKY rats (Panico, Luo et al. 2009), suggesting the effects of ROS might be associated with pathological conditions, such as hypertension. Interestingly,

Ortiz and Garvin have reported that ROS (superoxide) stimulate sodium reabsorption in the thick ascending limb of loop of Henle (THAL) and the collecting duct (Ortiz and

Garvin 2002; Silva, Ortiz et al. 2006). The above experiment evidence indicates that elevated ROS levels within proximal tubules might serve an important negative feedback function, promoting excretion of excess sodium and fluid for maintaining body fluid

− balance. Besides, in the outer medullar, O2· production in medullary thick ascending

− limb (mTAL) via the NAD(P)H oxidase pathway and interactions of O2· and nitric oxide

(NO) determine the effectiveness of in situ free radical cross-talk between the mTAL and the vasa recta in terms of regulation of medullar blood flow (Mori and Cowley 2003).

In a word, fluid reabsorption is regulated by redox balance in renal proximal tubules. ROS generation from either high salt or high levels of glucose or ANG II inhibit the Na+, K+-ATPase, apical NHE3 and sodium/glucose transporter and then promote renal proximal tubular sodium and fluid excretion under certain circumstances (Johns,

O'Shaughnessy et al. ; Zhang, Imam et al. 2002; Han, Lee et al. 2005).

Taken together, there might be some cross-talk between Na/K-ATPase/c-Src signaling and ROS generation in terms of renal sodium handling (Liu, Kennedy et al.) based on the following evidence : (1) we have demonstrated that cardiotonic steroids such as ouabain decrease renal proximal tubular Na+ reabsorption by the redistribution of

Na/K-ATPase and NHE3 through Na/K-ATPase/c-Src signaling (Liu, Kesiry et al. 2004;

Liu, Liang et al. 2005; Liu and Shapiro 2007). (2) Activation of Na/K-ATPase/c-Src signaling stimulates mitochondrial ROS production. (3) c-Src as initiating step of Na/K-

26 ATPase signaling, is also redox-sensitive and required for ROS generation (derived from

NADPH oxidases) (Seshiah, Weber et al. 2002; Touyz, Yao et al. 2003). (4) NADPH oxidase-derived ROS production reduces renal proximal tubular sodium reabsorption

(Panico, Luo et al. 2009).

Figure 1-8: Schematic illustration of the effects of ROS and Na/K-ATPase signaling on RPT sodium reabsorption (Liu, Kennedy et al. 2012). CTS, cardiotonic steroids; RPT, renal proximal tubules.

6.3.2 ROS and blood pressure regulation Oxidative stress is both a cause and consequence of hypertension (Kitiyakara,

Chabrashvili et al. 2003; Touyz 2004; Wilcox 2005; Vaziri and Rodriguez-Iturbe 2006;

Welch 2006).

A large body of evidence supports a role for ROS in the pathogenesis of hypertension. Increased ROS generation precedes development of hypertension, suggesting that ROS participate in the development and maintenance of hypertension

(Kitiyakara and Wilcox 1998; Houston 2005). In experimental hypertension including

- Dahl salt-sensitive hypertension, oxidative stress markers such as concentrations of O2

27 and H2O2, and activation of NADPH oxidase and xanthine oxidase are increased, whereas

levels of NO and antioxidant (SOD) are reduced (Kitiyakara, Chabrashvili et al.

2003; Touyz and Schiffrin 2004). In human, clinical studies have demonstrated that hypertensive patients produce excessive amounts of ROS (Lacy, Kailasam et al. 2000;

Fortuno, Olivan et al. 2004) and have reduced levels of antioxidant enzymes (SOD, glutathione peroxidase and catalase) (Saez, Tormos et al. 2004; Simic, Mimic-Oka et al.

2006). Based on the above experimental evidence and clinical studies, current therapeutic approaches that target ROS in the treatment of hypertension include the following mechanisms: (1) to increase antioxidant capacity through diet or supplementation, (2) to

- reduce ROS generation by decreasing activity of O2 generating enzymes, (3) and to

increase NO bioavailability(Hamilton, Miller et al. 2004; Touyz and Schiffrin 2004).

Antioxidant heme oxygenase (HO, encoded by HMOX genes) is the rate limiting

enzyme in heme degradation. Heme is a stable prosthetic group of a variety of enzymes.

A Fe2+ atom in its center of a protoporphyrin IX ring can be oxidized by hydrogen

peroxide to produce highly toxic hydroxyl radicals (HJH 1894). So it might be a

ubiquitous cytotoxic molecule involved in the pathogenesis of a broad spectrum of

diseases. HO can catalyze free heme to produce equimolar amounts of liable Fe (stored

by the ferritin H chain, FtH), carbon monoxide (CO), and biliverdin. Biliverdin is

subsequently converted to endogenous antioxidant bilirubin by biliverdin reductase

(Ndisang, Tabien et al. 2004; Abraham and Kappas 2005). CO like NO, signals and

augment NO levels to favor vasodilation and saliuresis. Bilirubin, which may enhance

- NO levels by reducing its rate of consumption by O2 , is an effective ROS scavenger

(Pallone 2007). All three end products of heme catabolism are cytoprotective due to their

28 potent antioxidant properties and antiapoptotic effects (Stocker, Yamamoto et al. 1987;

Abraham, Cao et al. 2009). Humans and rodents have two HO isoenzymes, namely HO-1

(32kDa) and HO-2 (36kDa) encoded by the HMOX1 and HMOX2 genes respectively.

HO-1 expression is induced ubiquitously in response to oxidative stress, whereas HO-2 is constitutively expressed and not inducible.

It has been shown that induction of HO-1 improves vascular function and ameliorates both genetic and experimental hypertension (Sacerdoti, Escalante et al. 1989;

Levere, Martasek et al. 1990; Sabaawy, Zhang et al. 2001; Yang, Quan et al. 2004;

Abraham and Kappas 2005; Botros, Schwartzman et al. 2005; Wang, Shamloul et al.

2006). Inhibition of HO-1 increases mean arterial pressure in Sprague-Dawley rats

(Johnson, Lavesa et al. 1995; Hirakawa and Hayashida 2006) and SHR rats (Johnson,

Lavesa et al. 1996). However, most current studies focus on vasculature. Given the crucial role of the kidneys in the long-term control of arterial blood pressure, it is imperative to determine whether and how local HOs in the kidneys, especial renal proximal tubules, participate in the regulation of the renal sodium excretion and arterial blood pressure. Li et al demonstrate that inhibition of renal medullary HO induces hypertension and increased salt sensitivity of arterial blood pressure through blunted natriuretic response to the elevation of renal perfusion pressure (Zou, Billington et al.

2000; Li, Yi et al. 2007; Pallone 2007). Increasingly, considerable attention has been paid mechanisms of pressure natriuresis because it favors amelioration of high blood pressure through reduction of extracellular fluid volume.

As discussed above (6.3.1.2), there might be some cross-talk between Na/K-

ATPase/c-Src signaling and ROS generation in terms of renal sodium handling (Liu,

29 Kennedy et al.). Binding of cardiotonic steroids such as ouabain to Na/K-ATPase decreases renal proximal tubular Na+ reabsorption by the redistribution of Na/K-ATPase and NHE3 through Na/K-ATPase/c-Src signaling (Liu, Kesiry et al. 2004; Liu, Liang et al. 2005; Liu and Shapiro 2007). Activation of Na/K-ATPase/c-Src signaling stimulates mitochondrial ROS production. c-Src as initiating step of Na/K-ATPase signaling, is also redox-sensitive and required for ROS generation (derived from NADPH oxidases)

(Seshiah, Weber et al. 2002; Touyz, Yao et al. 2003). NADPH oxidase-derived ROS production reduces renal proximal tubular sodium reabsorptiotn (Panico, Luo et al. 2009).

Together with the roles of renal medullar HO in regulation of salt and water excretion by the kidney, we propose a network among the Na/K-ATPase/c-Src signaling, NADPH oxidase-derived ROS generation and HO-1 that will favor understanding the ROS functional roles in renal proximal tubular sodium handling.

Although antioxidants exhibit a beneficial and protective effect on blood pressure in various animal models of hypertension, findings from clinical trials have been conflicting (reviewed in (Touyz 2004; Munzel, Gori et al. 2010). Some large trials including the Dietary Approaches to Stop Hypertension (DASH) have demonstrated that lower blood pressure associated with reduced dietary salt intake may be related to reductions in oxidative stress (Appel, Moore et al. 1997; Miller, Appel et al. 1998;

Sacks, Svetkey et al. 2001; John, Ziebland et al. 2002). However, antioxidant supplementation presently might not be recommended for the prevention and treatment of hypertension due to less evidence to prove the benefits from the routine use of antioxidants. Moreover, excessive antioxidant supplementation might be even dangerous (Huang, Caballero et al. 2006) due to the possible ‘over-antioxidant-

30 buffering’ effect. In this setting, excess antioxidants might become pro-oxidants (by

providing H+) if they cannot promptly be removed by the following anti-oxidant in the

biological anti-oxidant chain. Thus, it appears that the balance of the ROS status, within

a physiological range, may be more important to maintain a beneficial ROS signaling

(Liu, Kennedy et al.).

ROS is an appealing candidate target due to representing a common mechanism

leading from multiple risk factors to disease. Given the pivotal role of kidney in the

regulation of blood pressure, further studies, both in animal and humans, are needed in

order to achieve a better understanding of the role of ROS in the pathophysiology of

hypertension. Based on the literatures and our studies, this dissertation proposes: (1) ouabain stimulates trafficking regulation of the Na/K-ATPase and NHE3 in renal proximal tubule cells, (2) impairment of Na/K-ATPase signaling in renal proximal tubule contributes to Dahl salt-sensitive hypertension, (3) ROS are implicated in the modulation of the Na/K-ATPAse signaling and renal proximal tubular sodium handling. With greater insights and understanding of these processes regulating renal sodium handling and identification of molecular pathway that tip the equilibrium to states of oxidative stress which cause high blood pressure, the target therapies might be more effective in the prevention and treatment of hypertension.

31 References for literature review

Abraham, N. G., J. Cao, et al. (2009). "Heme oxygenase: the key to renal function

regulation." Am J Physiol Renal Physiol 297(5): F1137-52.

Abraham, N. G. and A. Kappas (2005). "Heme oxygenase and the cardiovascular-renal

system." Free Radic Biol Med 39(1): 1-25.

Albers, R. W. (1967). "Biochemical aspects of active transport." Annu Rev Biochem 36:

727-56.

Alexander, R. T. and S. Grinstein (2006). "Na+/H+ exchangers and the regulation of

volume." Acta Physiol (Oxf) 187(1-2): 159-67.

Alexander, R. T. and S. Grinstein (2009). "Tethering, recycling and activation of the

epithelial sodium-proton exchanger, NHE3." J Exp Biol 212(Pt 11): 1630-7.

Amemiya, M., J. Loffing, et al. (1995). "Expression of NHE-3 in the apical membrane of

rat renal proximal tubule and thick ascending limb." Kidney Int 48(4): 1206-15.

Anderson, D. E., O. V. Fedorova, et al. (2008). "Endogenous sodium pump inhibitors and

age-associated increases in salt sensitivity of blood pressure in normotensives."

Am J Physiol Regul Integr Comp Physiol 294(4): R1248-54.

Appel, L. J., T. J. Moore, et al. (1997). "A clinical trial of the effects of dietary patterns

on blood pressure. DASH Collaborative Research Group." N Engl J Med 336(16):

1117-24.

Axelsen, K. B. and M. G. Palmgren (1998). "Evolution of substrate specificities in the P-

type ATPase superfamily." J Mol Evol 46(1): 84-101.

Babior, B. M. (2004). "NADPH oxidase." Curr Opin Immunol 16(1): 42-7.

32 Bagrov, A. Y., N. I. Agalakova, et al. (2009). "Endogenous cardiotonic steroids and

differential patterns of sodium pump inhibition in NaCl-loaded salt-sensitive and

normotensive rats." Am J Hypertens 22(5): 559-63.

Bagrov, A. Y. and O. V. Fedorova (1998). "Effects of two putative endogenous digitalis-

like factors, marinobufagenin and ouabain, on the Na+, K+-pump in human

mesenteric arteries." J Hypertens 16(12 Pt 2): 1953-8.

Bagrov, A. Y., O. V. Fedorova, et al. (1996). "Plasma marinobufagenin-like and ouabain-

like immunoreactivity during saline volume expansion in anesthetized dogs."

Cardiovasc Res 31(2): 296-305.

Bagrov, A. Y. and J. I. Shapiro (2008). "Endogenous digitalis: pathophysiologic roles and

therapeutic applications." Nat Clin Pract Nephrol 4(7): 378-92.

Bagrov, A. Y., J. I. Shapiro, et al. (2009). "Endogenous cardiotonic steroids: physiology,

pharmacology, and novel therapeutic targets." Pharmacol Rev 61(1): 9-38.

Berlett, B. S. and E. R. Stadtman (1997). "Protein oxidation in aging, disease, and

oxidative stress." J Biol Chem 272(33): 20313-6.

Biemesderfer, D., J. Pizzonia, et al. (1993). "NHE3: a Na+/H+ exchanger isoform of

renal brush border." Am J Physiol 265(5 Pt 2): F736-42.

Blanco, G., J. C. Koster, et al. (1995). "Kinetic properties of the alpha 2 beta 1 and alpha

2 beta 2 isozymes of the Na,K-ATPase." Biochemistry 34(1): 319-25.

Blanco, G. and R. W. Mercer (1998). "Isozymes of the Na-K-ATPase: heterogeneity in

structure, diversity in function." Am J Physiol 275(5 Pt 2): F633-50.

Bokoch, G. M. and T. Zhao (2006). "Regulation of the phagocyte NADPH oxidase by

Rac GTPase." Antioxid Redox Signal 8(9-10): 1533-48.

33 Boldyrev, A., E. Bulygina, et al. (2003). "Na/K-ATPase regulates intracellular ROS level

in cerebellum neurons." Ann N Y Acad Sci 986: 519-21.

Boldyrev, A. and E. Kurella (1996). "Mechanism of oxidative damage of dog kidney

Na/K-ATPase." Biochem Biophys Res Commun 222(2): 483-7.

Boldyrev, A. A., E. R. Bulygina, et al. (1995). "[The effect of hydrogen peroxide and

hypochlorite on brain Na,K-ATPase activity]." Biokhimiia 60(10): 1688-96.

Botros, F. T., M. L. Schwartzman, et al. (2005). "Increase in heme oxygenase-1 levels

ameliorates renovascular hypertension." Kidney Int 68(6): 2745-55.

Bricker, N. S. (1967). "The control of sodium excretion with normal and reduced nephron

populations. The pre-eminence of third factor." Am J Med 43(3): 313-21.

Buckalew, V. M., Jr., F. J. Martinez, et al. (1970). "The effect of dialysates and

ultrafiltrates of plasma of saline-loaded dogs on toad bladder sodium transport." J

Clin Invest 49(5): 926-35.

Burritt, J. B., M. T. Quinn, et al. (1995). "Topological mapping of cytochrome

b epitopes with phage-display libraries." J Biol Chem 270(28): 16974-80.

Cai, H., L. Wu, et al. (2008). "Regulation of apical NHE3 trafficking by ouabain-induced

activation of the basolateral Na+-K+-ATPase receptor complex." Am J Physiol

Cell Physiol 294(2): C555-63.

Chevion, M., E. Berenshtein, et al. (2000). "Human studies related to protein oxidation:

protein carbonyl content as a marker of damage." Free Radic Res 33 Suppl: S99-

108.

Chow, C. W., S. Khurana, et al. (1999). "The epithelial Na(+)/H(+) exchanger, NHE3, is

internalized through a clathrin-mediated pathway." J Biol Chem 274(53): 37551-8.

34 Chow, D. C. and J. G. Forte (1995). "Functional significance of the beta-subunit for

heterodimeric P-type ATPases." J Exp Biol 198(Pt 1): 1-17.

Collazo, R., L. Fan, et al. (2000). "Acute regulation of Na+/H+ exchanger NHE3 by

parathyroid hormone via NHE3 phosphorylation and -dependent

endocytosis." J Biol Chem 275(41): 31601-8.

Cornelius, F. and Y. A. Mahmmoud (2003). "Functional modulation of the sodium pump:

the regulatory proteins "Fixit"." News Physiol Sci 18: 119-24.

Crajoinas, R. O., L. M. Lessa, et al. "Posttranslational mechanisms associated with

reduced NHE3 activity in adult vs. young prehypertensive SHR." Am J Physiol

Renal Physiol 299(4): F872-81.

Curtin, J. F., M. Donovan, et al. (2002). "Regulation and measurement of oxidative stress

in apoptosis." J Immunol Methods 265(1-2): 49-72.

D'Souza, S., A. Garcia-Cabado, et al. (1998). "The epithelial sodium-hydrogen antiporter

Na+/H+ exchanger 3 accumulates and is functional in recycling endosomes." J

Biol Chem 273(4): 2035-43.

Dada, L. A. and J. I. Sznajder (2003). "Mechanisms of pulmonary edema clearance

during acute hypoxemic respiratory failure: role of the Na,K-ATPase." Crit Care

Med 31(4 Suppl): S248-52.

De Wardener, H. E., I. H. Mills, et al. (1961). "Studies on the efferent mechanism of the

sodium diuresis which follows the administration of intravenous saline in the

dog." Clin Sci 21: 249-58.

Dobrota, D., M. Matejovicova, et al. (1999). "Na/K-ATPase under oxidative stress:

molecular mechanisms of injury." Cell Mol Neurobiol 19(1): 141-9.

35 Donowitz, M., S. Mohan, et al. (2009). "NHE3 regulatory complexes." J Exp Biol 212(Pt

11): 1638-46.

Dostanic-Larson, I., J. W. Van Huysse, et al. (2005). "The highly conserved cardiac

glycoside binding site of Na,K-ATPase plays a role in blood pressure regulation."

Proc Natl Acad Sci U S A 102(44): 15845-50.

Eakle, K. A., R. M. Lyu, et al. (1995). "The influence of beta subunit structure on the

interaction of Na+/K(+)-ATPase complexes with Na+. A chimeric beta subunit

reduces the Na+ dependence of phosphoenzyme formation from ATP." J Biol

Chem 270(23): 13937-47.

England, K., C. O'Driscoll, et al. (2004). "Carbonylation of glycolytic proteins is a key

response to drug-induced oxidative stress and apoptosis." Cell Death Differ 11(3):

252-60.

Fedorova, O. V., N. I. Agalakova, et al. (2005). "Brain ouabain stimulates peripheral

marinobufagenin via angiotensin II signalling in NaCl-loaded Dahl-S rats." J

Hypertens 23(8): 1515-23.

Fedorova, O. V., P. A. Doris, et al. (1998). "Endogenous marinobufagenin-like factor in

acute plasma volume expansion." Clin Exp Hypertens 20(5-6): 581-91.

Fedorova, O. V., N. I. Kolodkin, et al. (2001). "Marinobufagenin, an endogenous alpha-1

sodium pump ligand, in hypertensive Dahl salt-sensitive rats." Hypertension 37(2

Part 2): 462-6.

Fedorova, O. V., E. G. Lakatta, et al. (2000). "Endogenous Na,K pump ligands are

differentially regulated during acute NaCl loading of Dahl rats." Circulation

102(24): 3009-14.

36 Fedorova, O. V., M. I. Talan, et al. (2002). "Endogenous ligand of alpha(1) sodium pump,

marinobufagenin, is a novel mediator of sodium chloride--dependent

hypertension." Circulation 105(9): 1122-7.

Fedorova, O. V., I. A. Zhuravin, et al. (2007). "Intrahippocampal microinjection of an

exquisitely low dose of ouabain mimics NaCl loading and stimulates a

bufadienolide Na/K-ATPase inhibitor." J Hypertens 25(9): 1834-44.

Figtree, G. A., C. C. Liu, et al. (2009). "Reversible oxidative modification: a key

mechanism of Na+-K+ pump regulation." Circ Res 105(2): 185-93.

Finkel, T. and N. J. Holbrook (2000). "Oxidants, oxidative stress and the biology of

ageing." Nature 408(6809): 239-47.

Fisher, K. A., S. H. Lee, et al. (2001). "Regulation of proximal tubule sodium/hydrogen

antiporter with chronic volume contraction." Am J Physiol Renal Physiol 280(5):

F922-6.

Fontayne, A., P. M. Dang, et al. (2002). "Phosphorylation of p47phox sites by PKC alpha,

beta II, delta, and zeta: effect on binding to p22phox and on NADPH oxidase

activation." Biochemistry 41(24): 7743-50.

Fortuno, A., S. Olivan, et al. (2004). "Association of increased phagocytic NADPH

oxidase-dependent superoxide production with diminished nitric oxide generation

in essential hypertension." J Hypertens 22(11): 2169-75.

Geering, K. (2001). "The functional role of beta subunits in oligomeric P-type ATPases."

J Bioenerg Biomembr 33(5): 425-38.

Geering, K. (2006). "FXYD proteins: new regulators of Na-K-ATPase." Am J Physiol

Renal Physiol 290(2): F241-50.

37 Geering, K. (2008). "Functional roles of Na,K-ATPase subunits." Curr Opin Nephrol

Hypertens 17(5): 526-32.

Geiszt, M., J. B. Kopp, et al. (2000). "Identification of renox, an NAD(P)H oxidase in

kidney." Proc Natl Acad Sci U S A 97(14): 8010-4.

Graham, K. A., M. Kulawiec, et al. (2010). "NADPH oxidase 4 is an oncoprotein

localized to mitochondria." Cancer Biol Ther 10(3): 223-31.

Griendling, K. K. (2006). "NADPH oxidases: new regulators of old functions." Antioxid

Redox Signal 8(9-10): 1443-5.

Guyton, A. C. (1961). "Physiologic regulation of arterial pressure." Am J Cardiol 8: 401-

7.

Guyton, A. C. and T. G. Coleman (1999). "Quantitative analysis of the pathophysiology

of hypertension. 1969." J Am Soc Nephrol 10(10): 2248-58.

Halliwell, B. and J. M. Gutteridge (1990). "Role of free radicals and catalytic metal ions

in human disease: an overview." Methods Enzymol 186: 1-85.

Hamilton, C. A., W. H. Miller, et al. (2004). "Strategies to reduce oxidative stress in

cardiovascular disease." Clin Sci (Lond) 106(3): 219-34.

Hamlyn, J. M., M. P. Blaustein, et al. (1991). "Identification and characterization of a

ouabain-like compound from human plasma." Proc Natl Acad Sci U S A 88(14):

6259-63.

Han, H. J., Y. J. Lee, et al. (2005). "High glucose-induced oxidative stress inhibits

Na+/glucose cotransporter activity in renal proximal tubule cells." Am J Physiol

Renal Physiol 288(5): F988-96.

38 Hirakawa, H. and Y. Hayashida (2006). "Autonomic cardiovascular responses to heme

oxygenase inhibition in conscious rats." Hypertension 48(6): 1124-9.

HJH, F. (1894). "Oxidation of tartaric acid in presence of iron." J. Chem. Soc. 65: 899-

910.

Houston, M. C. (2005). "Nutraceuticals, vitamins, antioxidants, and minerals in the

prevention and treatment of hypertension." Prog Cardiovasc Dis 47(6): 396-449.

Hu, M. C., L. Fan, et al. (2001). "Dopamine acutely stimulates Na+/H+ exchanger

(NHE3) endocytosis via clathrin-coated vesicles: dependence on protein kinase

A-mediated NHE3 phosphorylation." J Biol Chem 276(29): 26906-15.

Huang, H.-Y., B. Caballero, et al. (2006). "The Efficacy and Safety of Multivitamin and

Mineral Supplement Use To Prevent Cancer and Chronic Disease in Adults: A

Systematic Review for a National Institutes of Health State-of-the-Science

Conference." Annals of Internal Medicine 145(5): 372-385.

Huang, W. H., Y. Wang, et al. (1992). "(Na+ + K+)-ATPase: inactivation and

degradation induced by oxygen radicals." Int J Biochem 24(4): 621-6.

Huang, W. H., Y. Wang, et al. (1994). "Different sensitivities of the Na+/K(+)-ATPase

isoforms to oxidants." Biochim Biophys Acta 1190(1): 108-14.

Huang, Y. T., S. C. Chueh, et al. (2004). "Investigation of ouabain-induced anticancer

effect in human androgen-independent prostate cancer PC-3 cells." Biochem

Pharmacol 67(4): 727-33.

Jaisser, F., J. D. Horisberger, et al. (1992). "The beta subunit modulates potassium

activation of the Na-K pump." Ann N Y Acad Sci 671: 113-9.

39 Janecki, A. J., M. Janecki, et al. (2000). "Basic fibroblast growth factor stimulates surface

expression and activity of Na(+)/H(+) exchanger NHE3 via mechanism involving

phosphatidylinositol 3-kinase." J Biol Chem 275(11): 8133-42.

John, J. H., S. Ziebland, et al. (2002). "Effects of fruit and vegetable consumption on

plasma antioxidant concentrations and blood pressure: a randomised controlled

trial." Lancet 359(9322): 1969-74.

Johns, E. J., B. O'Shaughnessy, et al. "Impact of elevated dietary sodium intake on

NAD(P)H oxidase and SOD in the cortex and medulla of the rat kidney." Am J

Physiol Regul Integr Comp Physiol 299(1): R234-40.

Johnson, R. A., M. Lavesa, et al. (1995). "A heme oxygenase product, presumably carbon

monoxide, mediates a vasodepressor function in rats." Hypertension 25(2): 166-9.

Johnson, R. A., M. Lavesa, et al. (1996). "Heme oxygenase substrates acutely lower

blood pressure in hypertensive rats." Am J Physiol 271(3 Pt 2): H1132-8.

Jorgensen, P. L. and J. P. Andersen (1988). "Structural basis for E1-E2 conformational

transitions in Na,K-pump and Ca-pump proteins." J Membr Biol 103(2): 95-120.

Kajikawa, M., S. Fujimoto, et al. (2002). "Ouabain suppresses glucose-induced

mitochondrial ATP production and insulin release by generating reactive oxygen

species in pancreatic islets." Diabetes 51(8): 2522-9.

Kennedy, D. J., S. Vetteth, et al. (2006). "Central role for the cardiotonic steroid

marinobufagenin in the pathogenesis of experimental uremic cardiomyopathy."

Hypertension 47(3): 488-95.

40 Kitiyakara, C., T. Chabrashvili, et al. (2003). "Salt intake, oxidative stress, and renal

expression of NADPH oxidase and superoxide dismutase." J Am Soc Nephrol

14(11): 2775-82.

Kitiyakara, C. and C. S. Wilcox (1998). "Antioxidants for hypertension." Curr Opin

Nephrol Hypertens 7(5): 531-8.

Komiyama, Y., X. H. Dong, et al. (2005). "A novel endogenous digitalis, telocinobufagin,

exhibits elevated plasma levels in patients with terminal renal failure." Clin

Biochem 38(1): 36-45.

Krzesinski, J. M. and E. P. Cohen (2007). "Salt, the kidneys, and arterial hypertension."

Acta Clin Belg 62(5): 348-57.

Kurashima, K., E. Z. Szabo, et al. (1998). "Endosomal recycling of the Na+/H+

exchanger NHE3 isoform is regulated by the phosphatidylinositol 3-kinase

pathway." J Biol Chem 273(33): 20828-36.

Kurella, E. G., O. V. Tyulina, et al. (1999). "Oxidative resistance of Na/K-ATPase." Cell

Mol Neurobiol 19(1): 133-40.

Kuster, B., A. Shainskaya, et al. (2000). "A new variant of the gamma subunit of renal

Na,K-ATPase. Identification by mass spectrometry, antibody binding, and

expression in cultured cells." J Biol Chem 275(24): 18441-6.

Lacy, F., M. T. Kailasam, et al. (2000). "Plasma hydrogen peroxide production in human

essential hypertension: role of heredity, gender, and ethnicity." Hypertension

36(5): 878-84.

41 Lacy, F., D. T. O'Connor, et al. (1998). "Plasma hydrogen peroxide production in

hypertensives and normotensive subjects at genetic risk of hypertension." J

Hypertens 16(3): 291-303.

Lander, H. M. (1997). "An essential role for free radicals and derived species in signal

transduction." FASEB J 11(2): 118-24.

Laredo, J., J. R. Shah, et al. (1997). "Angiotensin II stimulates secretion of endogenous

ouabain from bovine adrenocortical cells via angiotensin type 2 receptors."

Hypertension 29(1 Pt 2): 401-7.

Ledoussal, C., J. N. Lorenz, et al. (2001). "Renal salt wasting in mice lacking NHE3

Na+/H+ exchanger but not in mice lacking NHE2." Am J Physiol Renal Physiol

281(4): F718-27.

Lees, G. J. (1991). "Inhibition of sodium-potassium-ATPase: a potentially ubiquitous

mechanism contributing to central nervous system neuropathology." Brain Res

Brain Res Rev 16(3): 283-300.

Levere, R. D., P. Martasek, et al. (1990). "Effect of heme arginate administration on

blood pressure in spontaneously hypertensive rats." J Clin Invest 86(1): 213-9.

Li, N., F. Yi, et al. (2007). "Role of renal medullary heme oxygenase in the regulation of

pressure natriuresis and arterial blood pressure." Hypertension 49(1): 148-54.

Liang, M., T. Cai, et al. (2006). "Functional characterization of Src-interacting Na/K-

ATPase using RNA interference assay." J Biol Chem 281(28): 19709-19.

Lichtstein, D., I. Gati, et al. (1993). "Identification of digitalis-like compounds in human

cataractous lenses." Eur J Biochem 216(1): 261-8.

42 Lingrel, J. B. and T. Kuntzweiler (1994). "Na+,K(+)-ATPase." J Biol Chem 269(31):

19659-62.

Lingrel, J. B., J. Orlowski, et al. (1990). "Molecular genetics of Na,K-ATPase." Prog

Nucleic Acid Res Mol Biol 38: 37-89.

Liu, J., D. J. Kennedy, et al. "Reactive Oxygen Species Modulation of Na/K-ATPase

Regulates Fibrosis and Renal Proximal Tubular Sodium Handling." Int J Nephrol

2012: 381320.

Liu, J., D. J. Kennedy, et al. (2012). "Reactive Oxygen Species Modulation of Na/K-

ATPase Regulates Fibrosis and Renal Proximal Tubular Sodium Handling "

International Journl of Nephrology.

Liu, J., D. J. Kennedy, et al. (2012). "Reactive Oxygen Species Modulation of Na/K-

ATPase Regulates Fibrosis and Renal Proximal Tubular Sodium Handling." Int J

Nephrol 2012: 381320.

Liu, J., R. Kesiry, et al. (2004). "Ouabain induces endocytosis of plasmalemmal Na/K-

ATPase in LLC-PK1 cells by a clathrin-dependent mechanism." Kidney Int 66(1):

227-41.

Liu, J., M. Liang, et al. (2005). "Ouabain-induced endocytosis of the plasmalemmal

Na/K-ATPase in LLC-PK1 cells requires caveolin-1." Kidney Int 67(5): 1844-54.

Liu, J., S. M. Periyasamy, et al. (2002). "Effects of cardiac glycosides on sodium pump

expression and function in LLC-PK1 and MDCK cells." Kidney Int 62(6): 2118-

25.

43 Liu, J. and J. I. Shapiro (2007). "Regulation of sodium pump endocytosis by cardiotonic

steroids: Molecular mechanisms and physiological implications."

Pathophysiology 14(3-4): 171-81.

Liu, J., J. Tian, et al. (2000). "Ouabain interaction with cardiac Na+/K+-ATPase initiates

signal cascades independent of changes in intracellular Na+ and Ca2+

concentrations." J Biol Chem 275(36): 27838-44.

Liu, J. and Z. J. Xie (2010). "The sodium pump and cardiotonic steroids-induced signal

transduction protein kinases and calcium-signaling microdomain in regulation of

transporter trafficking." Biochim Biophys Acta 1802(12): 1237-45.

Liu, J., Y. Yan, et al. "Impairment of Na/K-ATPase signaling in renal proximal tubule

contributes to Dahl salt-sensitive hypertension." J Biol Chem 286(26): 22806-13.

Loreaux, E. L., B. Kaul, et al. (2008). "Ouabain-Sensitive alpha1 Na,K-ATPase enhances

natriuretic response to saline load." J Am Soc Nephrol 19(10): 1947-54.

Lorenz, J. N., P. J. Schultheis, et al. (1999). "Micropuncture analysis of single-nephron

function in NHE3-deficient mice." Am J Physiol 277(3 Pt 2): F447-53.

Lutsenko, S. and J. H. Kaplan (1993). "An essential role for the extracellular domain of

the Na,K-ATPase beta-subunit in cation occlusion." Biochemistry 32(26): 6737-

43.

Lutsenko, S. and J. H. Kaplan (1995). "Organization of P-type ATPases: significance of

structural diversity." Biochemistry 34(48): 15607-13.

Manunta, P., B. P. Hamilton, et al. (2006). "Salt intake and depletion increase circulating

levels of endogenous ouabain in normal men." Am J Physiol Regul Integr Comp

Physiol 290(3): R553-9.

44 Manunta, P., P. Stella, et al. (1999). "Left ventricular mass, stroke volume, and ouabain-

like factor in essential hypertension." Hypertension 34(3): 450-6.

Martyn, K. D., L. M. Frederick, et al. (2006). "Functional analysis of Nox4 reveals

unique characteristics compared to other NADPH oxidases." Cell Signal 18(1):

69-82.

McDonough, A. A. "Mechanisms of proximal tubule sodium transport regulation that

link extracellular fluid volume and blood pressure." Am J Physiol Regul Integr

Comp Physiol 298(4): R851-61.

McKinnon, W., G. A. Lord, et al. (2003). "Circulating sodium pump inhibitors in five

volume-expanded humans." J Hypertens 21(12): 2315-21.

Mercer, R. W. (1993). "Structure of the Na,K-ATPase." Int Rev Cytol 137C: 139-68.

Miller, E. R., 3rd, L. J. Appel, et al. (1998). "Effect of dietary patterns on measures of

lipid peroxidation: results from a randomized clinical trial." Circulation 98(22):

2390-5.

Miller, G., N. Suzuki, et al. "Reactive oxygen species homeostasis and signalling during

drought and salinity stresses." Plant Cell Environ 33(4): 453-67.

Moe, O. W., A. Tejedor, et al. (1991). "Dietary NaCl modulates Na(+)-H+ antiporter

activity in renal cortical apical membrane vesicles." Am J Physiol 260(1 Pt 2):

F130-7.

Morduchowicz, G. A., D. Sheikh-Hamad, et al. (1989). "Increased Na+/H+ antiport

activity in the renal brush border membrane of SHR." Kidney Int 36(4): 576-81.

45 Mori, T. and A. W. Cowley, Jr. (2003). "Angiotensin II-NAD(P)H oxidase-stimulated

superoxide modifies tubulovascular nitric oxide cross-talk in renal outer medulla."

Hypertension 42(4): 588-93.

Morth, J. P., B. P. Pedersen, et al. (2007). "Crystal structure of the sodium-potassium

pump." Nature 450(7172): 1043-9.

Munzel, T., T. Gori, et al. (2010). "Is oxidative stress a therapeutic target in

cardiovascular disease?" Eur Heart J 31(22): 2741-8.

Ndisang, J. F., H. E. Tabien, et al. (2004). "Carbon monoxide and hypertension." J

Hypertens 22(6): 1057-74.

Nesher, M., M. Dvela, et al. (2009). "Physiological roles of endogenous ouabain in

normal rats." Am J Physiol Heart Circ Physiol 297(6): H2026-34.

Newman, R. A., P. Yang, et al. (2008). "Cardiac glycosides as novel cancer therapeutic

agents." Mol Interv 8(1): 36-49.

Ortiz, P. A. and J. L. Garvin (2002). "Superoxide stimulates NaCl absorption by the thick

ascending limb." Am J Physiol Renal Physiol 283(5): F957-62.

Oweis, S., L. Wu, et al. (2006). "Cardiac glycoside downregulates NHE3 activity and

expression in LLC-PK1 cells." Am J Physiol Renal Physiol 290(5): F997-1008.

Pallone, T. L. (2007). "Control of renal Na+ excretion by heme oxygenase."

Hypertension 49(1): 23-4.

Panico, C., Z. Luo, et al. (2009). "Renal proximal tubular reabsorption is reduced in adult

spontaneously hypertensive rats: roles of superoxide and Na+/H+ exchanger 3."

Hypertension 54(6): 1291-7.

46 Paravicini, T. M. and R. M. Touyz (2006). "Redox signaling in hypertension."

Cardiovasc Res 71(2): 247-58.

Paravicini, T. M. and R. M. Touyz (2008). "NADPH oxidases, reactive oxygen species,

and hypertension: clinical implications and therapeutic possibilities." Diabetes

Care 31 Suppl 2: S170-80.

Pedemonte, C. H. and J. H. Kaplan (1990). "Chemical modification as an approach to

elucidation of sodium pump structure-function relations." Am J Physiol 258(1 Pt

1): C1-23.

Periyasamy, S. M., J. Liu, et al. (2005). "Salt loading induces redistribution of the

plasmalemmal Na/K-ATPase in proximal tubule cells." Kidney Int 67(5): 1868-77.

Petrushanko, I., N. Bogdanov, et al. (2006). "Na-K-ATPase in rat cerebellar granule cells

is redox sensitive." Am J Physiol Regul Integr Comp Physiol 290(4): R916-25.

Post, R. L., C. Hegyvary, et al. (1972). "Activation by adenosine triphosphate in the

phosphorylation kinetics of sodium and potassium ion transport adenosine

." J Biol Chem 247(20): 6530-40.

Pressley, T. A. (1996). "Structure and function of the Na,K pump: ten years of molecular

biology." Miner Electrolyte Metab 22(5-6): 264-71.

Rasmussen, H. H., E. J. Hamilton, et al. (2010). "Reversible oxidative modification:

implications for cardiovascular physiology and pathophysiology." Trends

Cardiovasc Med 20(3): 85-90.

Rivera, J., C. G. Sobey, et al. (2010). "Nox isoforms in vascular pathophysiology:

insights from transgenic and knockout mouse models." Redox Rep 15(2): 50-63.

47 Rossi, G., P. Manunta, et al. (1995). "Immunoreactive endogenous ouabain in primary

aldosteronism and essential hypertension: relationship with plasma renin,

aldosterone and blood pressure levels." J Hypertens 13(10): 1181-91.

Sabaawy, H. E., F. Zhang, et al. (2001). "Human heme oxygenase-1 gene transfer lowers

blood pressure and promotes growth in spontaneously hypertensive rats."

Hypertension 38(2): 210-5.

Sacerdoti, D., B. Escalante, et al. (1989). "Treatment with tin prevents the development

of hypertension in spontaneously hypertensive rats." Science 243(4889): 388-90.

Sachs, G. and K. Munson (1991). "Mammalian phosphorylating ion-motive ATPases."

Curr Opin Cell Biol 3(4): 685-94.

Sacks, F. M., L. P. Svetkey, et al. (2001). "Effects on blood pressure of reduced dietary

sodium and the Dietary Approaches to Stop Hypertension (DASH) diet. DASH-

Sodium Collaborative Research Group." N Engl J Med 344(1): 3-10.

Saez, G. T., C. Tormos, et al. (2004). "Factors related to the impact of antihypertensive

treatment in antioxidant activities and oxidative stress by-products in human

hypertension." Am J Hypertens 17(9): 809-16.

Schoner, W. and G. Scheiner-Bobis (2005). "Endogenous cardiac glycosides: hormones

using the sodium pump as signal transducer." Semin Nephrol 25(5): 343-51.

Schoner, W. and G. Scheiner-Bobis (2007). "Endogenous and exogenous cardiac

glycosides and their mechanisms of action." Am J Cardiovasc Drugs 7(3): 173-89.

Schoner, W. and G. Scheiner-Bobis (2007). "Endogenous and exogenous cardiac

glycosides: their roles in hypertension, salt metabolism, and ." Am J

Physiol Cell Physiol 293(2): C509-36.

48 Schreck, C. and P. M. O'Connor "NAD(P)H oxidase and renal epithelial ion transport."

Am J Physiol Regul Integr Comp Physiol 300(5): R1023-9.

Schreck, C. and P. M. O'Connor (2011). "NAD(P)H oxidase and renal epithelial ion

transport." Am J Physiol Regul Integr Comp Physiol 300(5): R1023-9.

Schultheis, P. J., L. L. Clarke, et al. (1998). "Renal and intestinal absorptive defects in

mice lacking the NHE3 Na+/H+ exchanger." Nat Genet 19(3): 282-5.

Sen, C. K. and L. Packer (1996). "Antioxidant and redox regulation of gene

transcription." FASEB J 10(7): 709-20.

Seshiah, P. N., D. S. Weber, et al. (2002). "Angiotensin II stimulation of NAD(P)H

oxidase activity: upstream mediators." Circ Res 91(5): 406-13.

Shinoda, T., H. Ogawa, et al. (2009). "Crystal structure of the sodium-potassium pump at

2.4 A resolution." Nature 459(7245): 446-50.

Shiose, A., J. Kuroda, et al. (2001). "A novel superoxide-producing NAD(P)H oxidase in

kidney." J Biol Chem 276(2): 1417-23.

Silva, G. B., P. A. Ortiz, et al. (2006). "Superoxide stimulates NaCl absorption in the

thick ascending limb via activation of protein kinase C." Hypertension 48(3): 467-

72.

Simic, D. V., J. Mimic-Oka, et al. (2006). "Byproducts of oxidative protein damage and

antioxidant enzyme activities in plasma of patients with different degrees of

essential hypertension." J Hum Hypertens 20(2): 149-55.

Skou, J. C. (1957). "The influence of some cations on an adenosine triphosphatase from

peripheral nerves." Biochim Biophys Acta 23(2): 394-401.

49 Stadtman, E. R. and R. L. Levine (2000). "Protein oxidation." Ann N Y Acad Sci 899:

191-208.

Stocker, R., Y. Yamamoto, et al. (1987). "Bilirubin is an antioxidant of possible

physiological importance." Science 235(4792): 1043-6.

Sweadner, K. J. and E. Rael (2000). "The FXYD gene family of small ion transport

regulators or channels: cDNA sequence, protein signature sequence, and

expression." Genomics 68(1): 41-56.

Thevenod, F. and J. M. Friedmann (1999). "Cadmium-mediated oxidative stress in

kidney proximal tubule cells induces degradation of Na+/K(+)-ATPase through

proteasomal and endo-/lysosomal proteolytic pathways." FASEB J 13(13): 1751-

61.

Tian, J., T. Cai, et al. (2006). "Binding of Src to Na+/K+-ATPase forms a functional

signaling complex." Mol Biol Cell 17(1): 317-26.

Tian, J., J. Liu, et al. (2003). "Involvement of mitogen-activated protein kinases and

reactive oxygen species in the inotropic action of ouabain on cardiac myocytes. A

potential role for mitochondrial K(ATP) channels." Mol Cell Biochem 242(1-2):

181-7.

Tian, J. and Z. J. Xie (2008). "The Na-K-ATPase and calcium-signaling microdomains."

Physiology (Bethesda) 23: 205-11.

Touyz, R. M. (2004). "Reactive oxygen species, vascular oxidative stress, and redox

signaling in hypertension: what is the clinical significance?" Hypertension 44(3):

248-52.

50 Touyz, R. M. (2006). "Lipid rafts take center stage in endothelial cell redox signaling by

death receptors." Hypertension 47(1): 16-8.

Touyz, R. M., X. Chen, et al. (2002). "Expression of a functionally active gp91phox-

containing neutrophil-type NAD(P)H oxidase in smooth muscle cells from human

resistance arteries: regulation by angiotensin II." Circ Res 90(11): 1205-13.

Touyz, R. M. and E. L. Schiffrin (2004). "Reactive oxygen species in vascular biology:

implications in hypertension." Histochem Cell Biol 122(4): 339-52.

Touyz, R. M., G. Yao, et al. (2003). "c-Src induces phosphorylation and translocation of

p47phox: role in superoxide generation by angiotensin II in human vascular

smooth muscle cells." Arterioscler Thromb Vasc Biol 23(6): 981-7.

Toyoshima, C., R. Kanai, et al. (2011). "First crystal structures of Na+,K+-ATPase: new

light on the oldest ion pump." Structure 19(12): 1732-8.

Traylor, L. A. and P. R. Mayeux (1997). "Superoxide generation by renal proximal tubule

nitric oxide synthase." Nitric Oxide 1(5): 432-8.

Vaziri, N. D. and B. Rodriguez-Iturbe (2006). "Mechanisms of disease: oxidative stress

and inflammation in the pathogenesis of hypertension." Nat Clin Pract Nephrol

2(10): 582-93.

Vignais, P. V. (2002). "The superoxide-generating NADPH oxidase: structural aspects

and activation mechanism." Cell Mol Life Sci 59(9): 1428-59.

Wang, H., M. Haas, et al. (2004). "Ouabain assembles signaling cascades through the

caveolar Na+/K+-ATPase." J Biol Chem 279(17): 17250-9.

51 Wang, R., R. Shamloul, et al. (2006). "Sustained normalization of high blood pressure in

spontaneously hypertensive rats by implanted hemin pump." Hypertension 48(4):

685-92.

Wang, X., I. Armando, et al. (2009). "The regulation of proximal tubular salt transport in

hypertension: an update." Curr Opin Nephrol Hypertens 18(5): 412-20.

Welch, W. J. (2006). "Intrarenal oxygen and hypertension." Clin Exp Pharmacol Physiol

33(10): 1002-5.

Wilcox, C. S. (2005). "Oxidative stress and nitric oxide deficiency in the kidney: a

critical link to hypertension?" Am J Physiol Regul Integr Comp Physiol 289(4):

R913-35.

Wilcox, C. S. and D. Gutterman (2005). "Focus on oxidative stress in the cardiovascular

and renal systems." Am J Physiol Heart Circ Physiol 288(1): H3-6.

Wondrak, G. T., D. Cervantes-Laurean, et al. (2000). "Histone carbonylation in vivo and

in vitro." Biochem J 351 Pt 3: 769-77.

Xie, Z. (2003). "Molecular mechanisms of Na/K-ATPase-mediated ."

Ann N Y Acad Sci 986: 497-503.

Xie, Z. and A. Askari (2002). "Na(+)/K(+)-ATPase as a signal transducer." Eur J

Biochem 269(10): 2434-9.

Xie, Z. and T. Cai (2003). "Na+-K+--ATPase-mediated signal transduction: from protein

interaction to cellular function." Mol Interv 3(3): 157-68.

Xie, Z., M. Jack-Hays, et al. (1995). "Different oxidant sensitivities of the alpha 1 and

alpha 2 isoforms of Na+/K(+)-ATPase expressed in baculovirus-infected insect

cells." Biochem Biophys Res Commun 207(1): 155-9.

52 Xie, Z., P. Kometiani, et al. (1999). "Intracellular reactive oxygen species mediate the

linkage of Na+/K+-ATPase to hypertrophy and its marker genes in cardiac

myocytes." J Biol Chem 274(27): 19323-8.

Xie, Z. J., Y. H. Wang, et al. (1990). "Studies on the specificity of the effects of oxygen

metabolites on cardiac sodium pump." J Mol Cell Cardiol 22(8): 911-20.

Yang, J., P. H. Lane, et al. (2009). "PKC-dependent superoxide production by the renal

medullary thick ascending limb from diabetic rats." Am J Physiol Renal Physiol

297(5): F1220-8.

Yang, L., S. Quan, et al. (2004). "Heme oxygenase-1 gene expression modulates

angiotensin II-induced increase in blood pressure." Hypertension 43(6): 1221-6.

Yang, L. E., P. K. Leong, et al. (2007). "Reducing blood pressure in SHR with enalapril

provokes redistribution of NHE3, NaPi2, and NCC and decreases NaPi2 and ACE

abundance." Am J Physiol Renal Physiol 293(4): F1197-208.

Yang, L. E., M. B. Sandberg, et al. (2008). "Effects of dietary salt on renal Na+

transporter subcellular distribution, abundance, and phosphorylation status." Am J

Physiol Renal Physiol 295(4): F1003-16.

Yang, X., M. Amemiya, et al. (2000). "Acid incubation causes exocytic insertion of

NHE3 in OKP cells." Am J Physiol Cell Physiol 279(2): C410-9.

Yates, N. A. and J. G. McDougall (1995). "Effect of volume expansion on the natriuretic

response to ouabain infusion." Ren Physiol Biochem 18(6): 311-20.

Zhang, A. Y., F. Yi, et al. (2006). "Lipid raft clustering and redox signaling platform

formation in coronary arterial endothelial cells." Hypertension 47(1): 74-80.

53 Zhang, C., S. Z. Imam, et al. (2002). "Peroxynitrite and the regulation of Na(+),K(+)-

ATPase activity by angiotensin II in the rat proximal tubule." Nitric Oxide 7(1):

30-5.

Zhang, R., P. Harding, et al. (2009). "Isoforms and functions of NAD(P)H oxidase at the

macula densa." Hypertension 53(3): 556-63.

Zhang, X., S. C. Zhang, et al. (2011). "New insight into the molecular mechanisms of the

biological effects of DNA minor groove binders." PLoS One 6(10): e25822.

Zolotarjova, N., C. Ho, et al. (1994). "Different sensitivities of native and oxidized forms

of Na+/K(+)-ATPase to intracellular proteinases." Biochim Biophys Acta 1192(1):

125-31.

Zou, A. P., H. Billington, et al. (2000). "Expression and actions of heme oxygenase in the

renal medulla of rats." Hypertension 35(1 Pt 2): 342-7.

Zuo, L., M. Ushio-Fukai, et al. (2005). "Caveolin-1 is essential for activation of Rac1 and

NAD(P)H oxidase after angiotensin II type 1 receptor stimulation in vascular

smooth muscle cells: role in redox signaling and vascular hypertrophy."

Arterioscler Thromb Vasc Biol 25(9): 1824-30.

54 Chapter 2 – Manuscript 1

Title:

Ouabain-Stimulated Trafficking Regulation of the Na/K-ATPase and NHE3 in

Renal Proximal Tubule Cells

Authors:

Yanling Yan1,3, Steven Haller1, Anna Shapiro1, Nathan Malhotra1, Jiang Tian1,

Zijian Xie2,1, Deepak Malhotra1, and Joseph I. Shapiro1,2, Jiang Liu1

From the Departments of Medicine1 and Physiology and Pharmacology2, University

of Toledo College of Medicine, Toledo, OH; and Institute of Biomedical

Engineering3, Yanshan University, China.

Corresponding Author:

Jiang Liu, M.D., Ph.D. Department of Medicine, University of Toledo College of

Medicine, Health Science Campus, 3000 Arlington Avenue, Toledo, OH, 43614

Phone: (419) 383-3923

Email: [email protected]

FAX: (419) 383-6244

Published in Molecular and Cellular Biochemistry. 2012, # : #-#. (mcbi-5426)

55 2.1 Abstract

We have demonstrated that ouabain regulates protein trafficking of the

Na/K-ATPase α1 subunit and NHE3 (Na/H exchanger, isoform 3) via ouabain-activated Na/K-ATPase signaling in porcine LLC-PK1 cells. To investigate

whether this mechanism is species-specific, ouabain-induced regulation of the α1

subunit and NHE3 as well as transcellular 22Na+ transport were compared in three

renal proximal tubular cell lines (human HK-2, porcine LLC-PK1, and AAC-19

originated from LLC-PK1 in which the pig α1 was replaced by ouabain-resistant rat

α1). Ouabain inhibited transcellular 22Na+ transport due to an ouabain-induced

redistribution of the α1 subunit and NHE3. In LLC-PK1 cells, ouabain also inhibited

the endocytic recycling of internalized NHE3, but has no significant effect on

recycling of endocytosed α1 subunit. These data indicated that the ouabain-induced

redistribution of the α1 subunit and NHE3 is not a species-specific phenomenon, and

ouabain-activated Na/K-ATPase signaling influences NHE3 regulation.

Keywords: Ouabain, Na/K-ATPase signaling, Na/K-ATPase, NHE3, redistribution

56 2.2 Introduction

Renal sodium handling is a key determinant of long term regulation of blood

pressure (Guyton 1991; Stamler, Rose et al. 1991; Meneton, Jeunemaitre et al. 2005;

Haddy 2006). In the kidney, the renal proximal tubules (RPTs) are responsible for

more than 60% of the net tubular Na+ reabsorption, mainly through basolateral

Na/K-ATPase and apical NHE3. Endogenous CTS (cardiotonic steroids, also known

as digitalis-like substances), which were initially classified as specific inhibitors of

the Na/K-ATPase and now classified as a new family of steroid hormones, are

involved in regulation of blood pressure and renal sodium handling (Schoner and

Scheiner-Bobis 2008; Bagrov, Shapiro et al. 2009; Nesher, Dvela et al. 2009).

Circulating CTS are markedly increased under certain conditions such as salt

loading, volume expansion, renal insufficiency, and congestive heart failure (Lloyd,

Sandberg et al. 1992; Fedorova, Doris et al. 1998; Manunta, Hamilton et al. 2006).

The pathophysiological significance of endogenous CTS has been a subject of

debate since it was first proposed (Haddy and Overbeck 1976; Blaustein 1977; De

Wardener 1977). In essence, the Na/K-ATPase inhibitor (endogenous CTS) will rise

in response to either a defect in renal Na+ excretion or high salt intake. This increase,

while returning Na+ balance toward normal by increasing renal Na+ excretion, also

cause increases in blood pressure through acting on vascular Na/K-ATPase

(Blaustein, Zhang et al. 2009). Increases in endogenous CTS regulate both renal Na+ excretion and blood pressure through the Na/K-ATPase (Dostanic-Larson, Van

57 Huysse et al. 2005; Loreaux, Kaul et al. 2008; Blaustein, Zhang et al. 2009; Nesher,

Dvela et al. 2009; Liu, Yan et al. 2011).

In porcine RPT LLC-PK1 cells, we have shown that ouabain inhibits active

transepithelial 22Na+ transport (from apical to basolateral aspect) via protein trafficking regulation of the Na/K-ATPase and NHE3 (Liu, Kesiry et al. 2004; Cai

2008; Liu and Xie 2010), a process requiring ouabain-activated Na/K-ATPase signaling. This novel regulatory mechanism may contribute to CTS-induced natriuresis, especially in rodents expressing ouabain-resistant Na/K-ATPase α1 subunit (Liu, Yan et al. 2011). In the present study, we investigated if this regulatory mechanism is species-specific by characterizing the effect of ouabain on transcellular 22Na+ flux and redistribution of the Na/K-ATPase and NHE3.

Furthermore, we also investigate the endocytic recycling (reinsertion of endocytosed

protein back to plasma membrane) of internalized Na/K-ATPase and NHE3 in

LLC-PK1 cells.

2.3 Experimental Methods

2.3.1 Chemicals and Antibodies

All reagents, unless otherwise mentioned, were obtained from Sigma (St.

Louis, MO). Src kinase inhibitor PP2 was from CalBiochem (San Diego, CA).

EZ-Kink sulfo-NHS-ss-Biotin and ImmunoPure immobilized streptavidin-agarose

beads were obtained from Pierce Biotechnology (Rockford, IL). A rabbit polyclonal

antibody against a mixture of peptides from porcine NHE3 was prepared and affinity

58 purified (Oweis, Wu et al. 2006). Antibodies against Rab7, integrin-β1 and human

NHE3 were from Santa Cruz Biotechnology (Santa Cruz, CA). Monoclonal antibody against the Na/K-ATPase α1-subunit (clone α6F) was from the

Developmental Studies Hybridoma Bank at the University of Iowa (Iowa City, IA).

Monoclonal antibodies against NHE3 (clone 4F5) and early endosome antigen-1

(EEA-1) were from Millipore Chemicon (Temecula, CA). Radioactive rubidium

(86Rb+) and sodium (22Na+) were from DuPont NEN Life Science (Boston, MA).

2.3.2 Cell cultures

Human HK-2 cells and pig LLC-PK1 cells were obtained from the American

Type Culture Collection (Manassas, VA). AAC-19 cells were generated from

LLC-PK1 cells as we described previously (Liang, Cai et al. 2006). Briefly, the

ouabain-sensitive pig α1 in LLC-PK1 cells was knock-down by siRNA method. To

rescue the α1 knock-down cell with ouabain-resistant rat α1 (AAC-19 cells), the α1

siRNA targeted sequence was silently mutated to introduce rat α1 with rat α1

pRc/CMV-α1AAC expression vector. The expression of ouabain-insensitive rat α1

was selected with 3µM ouabain in culture medium since untransfected LLC-PK1

cells are very sensitive to ouabain. Cells were cultured in DMEM (Dulbecco's

modified Eagle's medium)/F-12 mixed medium (1:1, vol/vol) for HK-2 or DMEM

for LLC-PK1 and AAC-19, with 10% fetal bovine serum (FBS), 100 U/ml penicillin,

and 100 µg/ml streptomycin) in a 5% CO2-humidified incubator. Culture medium

was changed daily until confluency. LLC-PK1 cells and AAC-19 cells were

serum-starved (in serum-free DMEM medium) for 16-18 h before treatment, and

59 HK-2 cells were changed to medium containing 1% FBS for 16-18 h before treatment. In assays for active transcellular 22Na+ flux, cells were grown on

Transwell membrane support to form monolayer, and then treated with ouabain either in the basolateral or apical compartment. Both LLC-PK1 and AAC-19 cells can be easily grown to monolayers in DMEM medium with 10% FBS. While HK-2 cells are hard to form monolayer with ATCC-recommended Keratinocyte Serum

Free Medium, we found that HK-2 cells could be easily grown to monolayer in

DMEM/F-12 medium (with 10% FBS) without losing its ouabain sensitivity.

2.3.3 Isolation of early endosome (EE) and late endosome (LE) fractions

EE and LE fractions were fractionated by a sucrose flotation gradient technique as we previously described (Liu, Kesiry et al. 2004; Cai 2008). EE and LE fractions were identified with antibodies against EE marker protein EEA-1 and LE marker protein Rab7, respectively (Liu, Kesiry et al. 2004; Cai 2008). In comparison with whole cell lysates, more than a 10-fold enrichment of these marker proteins was observed in representative endosome fractions as we have previously shown (Liu,

Kesiry et al. 2004).

2.3.4 Cell surface biotinylation

Cell surface biotinylation was conducted as we described before (Liu, Kesiry et al. 2004; Cai 2008). Biotinylated proteins were pulled down with streptavidin-agarose beads, eluted with 2x Laemmli buffer (125 mM Tris-HCl, 20%

Glycerol, 4% SDS, 0.025% Bromophenol Blue, 10% 2-mercaptoethanol, pH 6.8) at

60 55°C waterbath for 30 minutes, resolved by 10% SDS-PAGE, and then

immunoblotted for the Na/K-ATPase α1 and NHE3. The same membrane was also

imunoblotted with antibody against integrin-β1 to serve as loading control as

described previously (Liu, Yan et al. 2011).

2.3.5 Ouabain-sensitive Na/K-ATPase activity assay (86Rb+ uptake)

For 86Rb+ uptake assay, cells were cultured in 12-well plates and treated with or without different concentrations of ouabain for 15min. Monensin (20 µM), a

Na+-clamping agent, was added to the medium prior to the assay to assure that the

maximal capacity of active uptake was measured (Haber, Pressley et al. 1987). 86Rb+ uptake was initiated by the addition of 1 µCi of 86Rb+ as tracer of K+ to each well,

and the reaction was stopped after 15 minutes by washing four times with ice-cold

86 + 0.1 M MgCl2. Trichloroacetic acid (TCA)-soluble Rb was extracted with 10%

TCA and counted. TCA-precipitated cellular protein content was determined and used to calibrate the 86Rb+ uptake. Data were expressed as the percentage of control

86Rb+ uptake.

2.3.6 NHE3 mediated active transepithelial 22Na+ flux and 22Na+ uptake

Active transepithelial 22Na+ flux (from apical to basolateral aspect of the

Transwell membrane supports) was performed on monolayers (grown on Costar

Transwell culture filter inserts, filter pore size: 0.4 µm, Costar, Cambridge, MA) as

described by Haggerty and colleagues (Haggerty, Agarwal et al. 1988). Briefly, after

ouabain treatment for 1h at the concentration indicated, both apical (upper) and

61 basolateral (lower) compartments were rinsed with ouabain-free DMEM (0% FBS

for LLC-PK1 and AAC-19 cells, and 1% FBS for HK-2 cells). 1 ml DMEM

containing 22Na+ (1 µCi/ml) was added to the apical compartment of a filter insert, and the basolateral compartment was filled with 1 ml of DMEM. After 1h, aliquots

were removed from the basolateral compartments for scintillation counting.

H+-driven 22Na+ uptake were determined as described by Soleimani and colleagues

(Soleimani, Watts et al. 1998). Briefly,the cells grown on 12-well plate were treated

with ouabain at the concentration indicated and then washed three times with the

Na+-free buffer (in mM, 140 N-methyl-D-glucammonium (NMDG+) Cl, 4 KCl, 2

MgCl2, 1 CaCl2, 10 HEPES, pH 7.4). The cells were then incubated for 10 min in the

+ + + same Na -free buffer in which 20 mM NMDG was replaced with 20 mM NH4 . The

+ + uptake was initiated by replacing the NH4 -containing buffer with Na -free buffer

containing 1 µCi/ml 22NaCl+. 22Na+ uptake was stopped after 30 min by washing four

times with ice-cold saline. Cell-associated radioactivity was extracted with 1 ml of 1

N sodium hydroxide, quantified by scintillation counting, and calibrated with protein

content. In both experimental settings, cells were pretreated with 50 µM amiloride to

inhibit amiloride-sensitive NHE1 activity.

2.3.7 Assessment of endocytic recycling of NHE3 and Na/K-ATPase α1 subunit

in LLC-PK1 cells

Endocytic recycling of α1 and NHE3 were assessed by the method described

by the Moe Laboratory (Klisic, Zhang et al. 2003). Briefly, two sets of LLC-PK1

cells were biotinylated and quenched at 4°C, and then treated with ouabain (100nM)

62 or vehicle (as control) for 1h at 37°C to induce redistribution of the Na/K-ATPase α1

and NHE3. After rinsing with ice-cold PBS-Ca-Mg (1x PBS with 0.1mM CaCl2 and

1mM MgCl2), un-internalized surface biotinylated proteins were cleaved with 50

mM glutathione-SH (GSH-SH, a reducing agent) at 4°C, and un-reacted free

GSH-SH was oxidized by incubation with 30mM iodoacetamide for 10min. At this

point, one set of cells was lysed with RIPA buffer to retrieve total internalized

intracellular biotinylated α1 and NHE3 with streptavidin-agarose beads. Other set of

cells was changed to serum-free DMEM medium and cultured at 37°C to permit

further trafficking for 2h with or without 100nM ouabain. Biotinylated proteins that

recycled back to cell surface (reinsertion) were cleaved by GSH-SH method again.

The remaining intracellular biotinylated proteins after reinsertion were retrieved

with streptavidin-agarose beads, which represent the internalized biotinylated α1 and NHE3 that were not reinserted. The difference of the total intracellular α1 and

NHE3, before and after reinsertion, represents the α1 and NHE3 that was

internalized and then reinserted (endocytic recycling).

2.3.8 Western blot

Equal amounts of total protein were resolved by 10% SDS-PAGE and

immunoblotted with indicated antibodies (with dilution of 1:2000 for the

Na/K-ATPase α1 subunit and 1:1000 dilution for NHE3, in 4% non-fat dry milk in

1x Tris-buffered saline with 0.1% Tween-20). The same membrane was also

imunoblotted with antibodies against integrin-β1 (for surface biotinylation), EEA-1

(for EE fraction) and Rab7 (for LE fraction) to serve as loading controls (data not

63 shown), respectively, as we previously described (Liu, Kesiry et al. 2004; Cai 2008;

Liu, Yan et al. 2011). Signal detection was performed with an enhanced

chemiluminescence super signal kit (Pierce, Rockford, IL). Multiple exposures were

analyzed to assure that the signals were within the linear range of the film. The signal density was determined using Molecular Analyst software (Bio-Rad, Hercules, CA).

2.3.9 Statistical analysis

Data were tested for normality (all data passed) and then subjected to parametric analysis. When more than two groups were compared, one-way ANOVA

was performed prior to the comparison of individual groups with an unpaired t-test.

Statistical significance was reported at the P < 0.05 and P < 0.01 levels. SPSS

software was used for all analysis (SPSS, Chicago, IL). Values are given as

mean±S.E.

2.4 Results

2.4.1 Ouabain-mediated inhibition of the Na/K-ATPase

Ouabain-induced inhibition of the Na/K-ATPase “ion-pumping” activity

(ouabain-sensitive 86Rb+ uptake) in these RPT cell lines is summarized in Fig. 2-1.

The IC50 values are consistent with the established differences of α1 ouabain

sensitivity amongst these species (see Discussion). In LLC-PK1 cells with IC50 at

1µM, 100nM ouabain is sufficient to activate the Na/K-ATPase signaling and consequent regulation of the Na/K-ATPase and NHE3 (Liu and Xie 2010).

64 According to the Na/K-ATPase α1 sensitivity to ouabain, we chose ouabain concentrations that are able to activate the Na/K-ATPase signaling for these three cell lines (10nM for HK-2, 100nM for LLC-PK1, and 10μM for AAC-19 cells) without significant inhibition of Na/K-ATPase activity. No significant effect on cell viability was observed when these cells were treated for 1h with ouabain concentrations used that was evaluated by Trypan blue exclusion.

2.4.2 Ouabain-mediated inhibition of transepithelial 22Na+ flux and

22Na+uptake

We have shown that ouabain inhibits transepithelial 22Na+ flux by activating

Na/K-ATPase signaling in LLC-PK1 cells (Liu, Kesiry et al. 2004; Cai 2008). To

assess if this effect is species-specific, we measured H+-driven 22Na+ uptake and

transepithelial 22Na+ flux in these three RPT cell lines. As shown in Fig. 2-2 and 2-3,

when ouabain was added in the basolateral aspect, ouabain inhibited 22Na+ uptake

(Fig. 2-2) and active transepithelial 22Na+ flux (Fig. 2-3) in both HK-2 and AAC-19 cells in the same manner as in LLC-PK1 cells. The effect of ouabain on 22Na+ flux

and NHE3 activity was largely blunted when these cells were pretreated with the Src

kinase inhibitor PP2 (1µM for 30 min, at 37 °C). PP2 alone did not show significant effect. No significant inhibition of NHE3 activity was observed in all three cell lines

when ouabain was added in the apical aspect (data not shown), suggesting that

ouabain-induced regulation of 22Na+ flux and NHE3 activity requires

ouabain-activated Na/K-ATPase signaling.

65 2.4.3 Ouabain-induced protein trafficking of Na/K-ATPase and NHE3

In LLC-PK1 cells, ouabain-induced inhibition of 22Na+ flux is largely due to

an ouabain-mediated redistribution of the Na/K-ATPase and NHE3 via

Na/K-ATPase signaling. To further test this regulatory mechanism, these three cell

lines were treated with or without ouabain to assess the ouabain-induced

redistribution. As shown in Fig. 2-4, the ouabain induced redistribution of the α1

subunit and NHE3 was caused by a decrease in cell surface α1 subunit and NHE3 in

both HK-2 and AAC-19 cells (as we previously reported in LLC-PK1 cells (Liu,

Kesiry et al. 2004; Cai 2008)). Pretreatment with PP2 abolished ouabain-induced

redistribution of the α1 subunit and NHE3 (data not shown). As shown in Table 2.1,

ouabain (1h) caused a dose-dependent reduction of cell surface α1 subunit and

NHE3. Furthermore, the ouabain-induced reduction of cell surface α1 subunit and

NHE3 was closely correlated to ouabain-induced inhibition of transcellular 22Na+ flux and NHE3 activity (Fig. 2-2 and 2-3).

2.4.4 Ouabain-mediated regulation of endocytic recycling of the Na/K-ATPase

α1 subunit and NHE3 in LLC-PK1 cells

Endocytic recycling is essential for maintaining the distinction between apical and basolateral membranes in polarized cells, even though the recycling pathways may be redundant (Maxfield and McGraw 2004). It has been shown that ouabain redistributes the Na/K-ATPase into both late endosomes and lysosomes

(Algharably, Owler et al. 1986), presumably for degradation. To explore the underlying mechanism, we used LLC-PK1 cells to assess endocytic recycling of the

66 α1 subunit and NHE3. As shown in Fig. 2-5A, GSH-SH (glutathione-SH, a reducing agent) was able to cleave over 90% of protein-bound biotin. Ouabain (100nM, 1h)

accumulated the α1 subunit (control 100±9.6% vs. ouabain 198.3±18.2, n=3, p<0.01)

and NHE3 (control 100±8.9% vs. ouabain 187.4±16.5, n=3, p<0.01) in intracellular

compartments (Fig. 2-5B) after cleavage of surface protein-bound biotin with

GSH-SH method. After 2h-period of recycling of internalized biotinylated proteins,

recycled biotinylated proteins were cleaved again with GSH-SH. The total

intracellular biotinylated α1 and NHE3 after recycling, which represent un-recycled

biotinylated α1 and NHE3, are shown in Fig. 2-5C. The reinsertion (endocytic

recycling) was presented as the difference of the total intracellular α1 and NHE3

before and after reinsertion (Fig. 2-5D). The present data indicate that ouabain

treatment not only induced NHE3 redistribution, but it also inhibited the endocytic

recycling of NHE3. Interestingly, the endocytic recycling of the α1 subunit was not

significantly affected by ouabain. These observations suggested that, while most of

the internalized α1 was destined for degradation, at least part of internalized NHE3

was recycled back to surface. Despite the lack of microvilli in

cultured renal proximal tubular cells (McDonough and Biemesderfer 2003), our

observation is reminiscent of the model of NHE3 moving along the microvilli

structure (Yang, Maunsbach et al. 2004).

To further explore the destination of the α1 subunit and NHE3, we determined

the protein content of these two transporters in EE and LE fractions in response to

ouabain. As shown in Fig. 2-6, ouabain treatment (100nM, 1h) stimulated

accumulation of both α1 and NHE3 in EE fractions. However, ouabain significantly

67 accumulated α1, but not NHE3 in LE fractions. The NHE3 protein content in LE fractions was not significantly increased after ouabain treatment, even in the presence of the lysosomotropic weak base agent chloroquine (0.2 mM, pretreated for

2h) which inhibits degradation by the lysosomal pathway. On the other hand, pretreatment with chloroquine caused a further accumulation of the α1 subunit in LE fractions in response to ouabain, suggesting that the endocytosed α1 subunit, but not

NHE3, is degraded through the LE-lysosome pathway.

2.5 Discussion

The renal tubular Na/K-ATPase comprises a final site for the regulation of renal sodium transport by many factors (Aperia 2001). Our recent work indicates that ouabain is one of these factors that act via a coordinated regulation of the

Na/K-ATPase and NHE3 through Na/K-ATPase signaling (Liu, Kesiry et al. 2004;

Cai 2008; Liu and Xie 2010; Liu, Yan et al. 2011). In the present study, we have investigated whether this ouabain mediated regulatory model is species-specific.

Our present study indicated that ouabain-induced regulation of the Na/K-ATPase and NHE3 is not species-specific. First, ouabain was able to inhibit the active transepithelial 22Na+ flux in these three cell lines. This effect was largely prevented by blocking c-Src activation with PP2 pretreatment. Secondly, ouabain-induced redistribution and reduction of the cell surface Na/K-ATPase and NHE3 contributed to the inhibition of active 22Na+ flux. Third, the effect of ouabain on active 22Na+ flux was only observed when ouabain was applied in the basolateral, but not in the apical

68 aspect. Fourth, the ouabain-induced reduction of the cell surface α1 subunit

contributed to the ouabain-induced inhibition of 22Na+ flux and 22Na+ uptake. Taken

together, these observations suggest that the ouabain-induced regulation of the

Na/K-ATPase and NHE3 is not species-specific, and may be explained if the

Na/K-ATPase is the functional receptor for ouabain-induced regulation.

Activation of c-Src is critical in the ouabain-activated Na/K-ATPase

signaling and redistribution of the Na/K-ATPase and NHE3 (Liu, Kesiry et al. 2004;

Liu, Liang et al. 2005; Oweis, Wu et al. 2006; Cai 2008; Liu and Xie 2010). When

compared with HK-2 and LLC-PK1 cells, a higher concentration of ouabain was

needed to regulate activity and redistribution of the Na/K-ATPase and NHE3 in

AAC-19 cells expressing native pig NHE3 and rat α1. This is consistent with the established differences in ouabain sensitivity of the α1 subunit as well as ouabain-stimulated c-Src activation between these two cell lines (Liang, Cai et al.

2006). It is well known that there are large differences in sensitivity of the

Na/K-ATPase to ouabain based on α isoforms and species (Sweadner 1989; Lingrel and Kuntzweiler 1994; Blanco and Mercer 1998). Specifically, the rodent α1 is far less sensitive than pig, dog, or human α1. Higher concentrations of ouabain were required to activate Na/K-ATPase signaling in rodents, compared to other species

(Liu, Tian et al. 2000; Aizman, Uhlen et al. 2001; Aydemir-Koksoy, Abramowitz et al. 2001; Haas, Wang et al. 2002; Liang, Cai et al. 2006). We have also shown that a higher concentration of ouabain (10µM) is needed to activate c-Src in isolated renal proximal tubules of Dahl salt-resistant rats (Liu, Yan et al. 2011). Most interestingly, different natriuretic responses were observed between transgenic mice expressing

69 ouabain-sensitive α1 and wild-type mice expressing ouabain-resistant α1(Loreaux,

Kaul et al. 2008) in which the ouabain binding site of the α1 subunit plays a critical

role (Lingrel 2010). As shown in Table 2.1, the ouabain sensitivity of the α1 subunit

influenced the ouabain-induced inhibition of 22Na+ flux as well as surface reduction of the α1 subunit and NHE3. The present study further suggests that species-specific

α1 sensitivity to ouabain might explain the species differences in ouabain-induced

natriuresis in vivo (Lloyd, Sandberg et al. 1992; Yates and McDougall 1993;

Loreaux, Kaul et al. 2008; Nesher, Dvela et al. 2009). Considering the high ouabain

sensitivity of human α1 subunit, this could be a explanation how pathophysiological

circulating CTS might affect renal sodium handling, especially in the view that

endogenous ouabain is a natriuretic hormone and has a physiological role in

controlling sodium homeostasis in normal rats (Nesher, Dvela et al. 2009).

After the redistribution of membrane proteins, subsequent intracellular trafficking differs among different endocytosed proteins. Receptor-mediated redistribution is believed to be an effective pathway to reduce cell surface signaling receptors. In our experimental settings, ouabain caused accumulation of the α1 subunit in LE fractions, but failed to affect its endocytic recycling (Fig. 2-5 and 6).

This is a reminiscence of the early observation that ouabain caused the redistribution of the Na/K-ATPase into late endosomes and lysosomes (Algharably, Owler et al.

1986), suggesting that the endocytosed Na/K-ATPase is most likely degraded in

LE/lysosome pathway. On the other hand, the cell surface expression of NHE3 is likely reduced by inhibition of the recycling of internalized NHE3 (Fig. 2-5 and 6).

However, the mechanism is not clear. In both cases, we cannot exclude the

70 possibility that both recycling and degradation were affected by ouabain, but the

overall observed effect was a balance between these two processes.

Renal Na+ reabsorption through NHE3 plays an important role in

salt-sensitivity, as well as in the development and control of sodium homeostasis and

blood pressure (Harris, Brenner et al. 1986; Kelly, Quinn et al. 1997; Schultheis,

Clarke et al. 1998; Lorenz, Schultheis et al. 1999). Recently, we have demonstrated

that impaired Na/K-ATPase-Src signaling contributes to salt sensitivity in Dahl rats

(Liu, Yan et al. 2011). This is consistent with the observation that a high salt diet

stimulates redistribution of RPT Na/K-ATPase and NHE3 (McDonough 2010).

Although the mechanisms are still being elucidated, accumulating evidence supports

the notion of coordinated regulation of the Na/K-ATPase and NHE3 (Liu and

Shapiro 2007). It appears that the pathways regulating the Na/K-ATPase and NHE3

are numerous and redundant, and CTS-induced coordinated regulation is one of the

pathways that occur in response to conditions that cause an increase in endogenous

CTS.

In summary, our data indicate that the Na/K-ATPase is the functional receptor for ouabain-induced regulation of the Na/K-ATPase and NHE3 (and thus transcellular Na+ transport), as we previously proposed (Liu and Xie 2010). This regulation is not species-specific, but the species-specific α1 ouabain sensitivity may

partially account for the species differences observed in ouabain-induced natriuresis

(Lloyd, Sandberg et al. 1992; Yates and McDougall 1993; Loreaux, Kaul et al. 2008;

Nesher, Dvela et al. 2009). In ouabain-induced trafficking regulation, endocytic recycling of internalized NHE3, but not the α1 subunit, was inhibited by ouabain.

71

Acknowledgments

The authors would like to thank Ms. Carol Woods her excellent help. Portions of this work were supported by National Institutes of Health Grants HL-105649 (to

J.T.), HL-109015 (to Z.X. and J.I.S.) and GM-78565 (to Z.X.).

72 2.6 References

Aizman, O., P. Uhlen, et al. (2001). "Ouabain, a steroid hormone that signals with slow

calcium oscillations." Proc Natl Acad Sci U S A 98(23): 13420-4.

Algharably, N., D. Owler, et al. (1986). "The rate of uptake of cardiac glycosides into

human cultured cells and the effects of chloroquine on it." Biochem Pharmacol

35(20): 3571-81.

Aperia, A. (2001). "Regulation of sodium/potassium ATPase activity: impact on salt

balance and vascular contractility." Curr Hypertens Rep 3(2): 165-71.

Aydemir-Koksoy, A., J. Abramowitz, et al. (2001). "Ouabain-induced signaling and

vascular smooth muscle cell proliferation." J Biol Chem 276(49): 46605-11.

Bagrov, A. Y., J. I. Shapiro, et al. (2009). "Endogenous cardiotonic steroids: physiology,

pharmacology, and novel therapeutic targets." Pharmacol Rev 61(1): 9-38.

Blanco, G. and R. W. Mercer (1998). "Isozymes of the Na-K-ATPase: heterogeneity in

structure, diversity in function." Am J Physiol 275(5 Pt 2): F633-50.

Blaustein, M. P. (1977). "Sodium ions, calcium ions, blood pressure regulation, and

hypertension: a reassessment and a hypothesis." Am J Physiol 232(5): C165-73.

Blaustein, M. P., J. Zhang, et al. (2009). "The pump, the exchanger, and endogenous

ouabain: signaling mechanisms that link salt retention to hypertension."

Hypertension 53(2): 291-8.

Cai, H., Wu, L., Qu, W., Malhotra, D., Xie, Z., Shapiro, JI, and Liu, J. (2008). "Regulation

of Apical NHE3 Trafficking by Ouabain-Induced Activation of Basolateral

Na/K-ATPase Receptor Complex." Am J Physiol Cell Physiol 294(2): C555-63.

73 De Wardener, H. E. (1977). "Natriuretic hormone." Clin Sci Mol Med 53(1): 1-8.

Dostanic-Larson, I., J. W. Van Huysse, et al. (2005). "The highly conserved cardiac

glycoside binding site of Na,K-ATPase plays a role in blood pressure regulation."

Proc Natl Acad Sci U S A 102(44): 15845-50.

Fedorova, O. V., P. A. Doris, et al. (1998). "Endogenous marinobufagenin-like factor in

acute plasma volume expansion." Clin Exp Hypertens 20(5-6): 581-91.

Guyton, A. C. (1991). "Blood pressure control--special role of the kidneys and body

fluids." Science 252(5014): 1813-6.

Haas, M., H. Wang, et al. (2002). "Src-mediated inter-receptor cross-talk between the

Na+/K+-ATPase and the epidermal growth factor receptor relays the signal from

ouabain to mitogen-activated protein kinases." J Biol Chem 277(21): 18694-702.

Haber, R. S., T. A. Pressley, et al. (1987). "Ionic dependence of active Na-K transport:

"clamping" of cellular Na+ with monensin." Am J Physiol 253(1 Pt 2): F26-33.

Haddy, F. J. (2006). "Role of dietary salt in hypertension." Life Sci 79(17): 1585-92.

Haddy, F. J. and H. W. Overbeck (1976). "The role of humoral agents in volume expanded

hypertension." Life Sci 19(7): 935-47.

Haggerty, J. G., N. Agarwal, et al. (1988). "Pharmacologically different Na/H antiporters

on the apical and basolateral surfaces of cultured porcine kidney cells (LLC-PK1)."

Proc Natl Acad Sci U S A 85(18): 6797-801.

Harris, R. C., B. M. Brenner, et al. (1986). "Sodium-hydrogen exchange and glucose

transport in renal microvillus membrane vesicles from rats with diabetes mellitus."

J Clin Invest 77(3): 724-33.

74 Kelly, M. P., P. A. Quinn, et al. (1997). "Activity and Expression of Na+-H+ Exchanger

Isoforms 1 and 3 in Kidney Proximal Tubules of Hypertensive Rats." Circ Res

80(6): 853-860.

Klisic, J., J. Zhang, et al. (2003). "Albumin regulates the Na+/H+ exchanger 3 in OKP

cells." J Am Soc Nephrol 14(12): 3008-16.

Liang, M., T. Cai, et al. (2006). "Functional Characterization of Src-interacting

Na/K-ATPase Using RNA Interference Assay." J Biol Chem 281(28):

19709-19719.

Lingrel, J. B. (2010). "The Physiological Significance of the Cardiotonic

Steroid/Ouabain-Binding Site of the Na,K-ATPase." Annual Review of Physiology

72: 395-412.

Lingrel, J. B. and T. Kuntzweiler (1994). "Na+,K(+)-ATPase." J Biol Chem 269(31):

19659-62.

Liu, J., R. Kesiry, et al. (2004). "Ouabain induces endocytosis of plasmalemmal

Na/K-ATPase in LLC-PK1 cells by a clathrin-dependent mechanism." Kidney Int

66(1): 227-41.

Liu, J., M. Liang, et al. (2005). "Ouabain-induced endocytosis of the plasmalemmal

Na/K-ATPase in LLC-PK1 cells requires caveolin-1." Kidney Int 67(5): 1844-54.

Liu, J. and J. I. Shapiro (2007). "Regulation of sodium pump endocytosis by cardiotonic

steroids: Molecular mechanisms and physiological implications." Pathophysiology

14(3-4): 171-81.

75 Liu, J., J. Tian, et al. (2000). "Ouabain interaction with cardiac Na+/K+-ATPase initiates

signal cascades independent of changes in intracellular Na+ and Ca2+

concentrations." J Biol Chem 275(36): 27838-44.

Liu, J. and Z. J. Xie (2010). "The sodium pump and cardiotonic steroids-induced signal

transduction protein kinases and calcium-signaling microdomain in regulation of

transporter trafficking." Biochim Biophys Acta 1802(12): 1237-45.

Liu, J., Y. Yan, et al. (2011). "Impairment of Na/K-ATPase Signaling in Renal Proximal

Tubule Contributes to Dahl Salt-sensitive Hypertension." J Biol Chem 286(26):

22806-13.

Lloyd, M. A., S. M. Sandberg, et al. (1992). "Role of renal Na+,K(+)-ATPase in the

regulation of sodium excretion under normal conditions and in acute congestive

heart failure." Circulation 85(5): 1912-7.

Loreaux, E. L., B. Kaul, et al. (2008). "Ouabain-Sensitive alpha1 Na,K-ATPase enhances

natriuretic response to saline load." J Am Soc Nephrol 19(10): 1947-54.

Lorenz, J. N., P. J. Schultheis, et al. (1999). "Micropuncture analysis of single-nephron

function in NHE3-deficient mice." Am J Physiol 277(3 Pt 2): F447-53.

Manunta, P., B. P. Hamilton, et al. (2006). "Salt intake and depletion increase circulating

levels of endogenous ouabain in normal men." Am J Physiol Regul Integr Comp

Physiol 290(3): R553-9.

Maxfield, F. R. and T. E. McGraw (2004). "Endocytic recycling." Nat Rev Mol Cell Biol

5(2): 121-32.

McDonough, A. A. (2010). "Mechanisms of proximal tubule sodium transport regulation

that link extracellular fluid volume and blood pressure." American Journal of

76 Physiology - Regulatory, Integrative and Comparative Physiology 298(4):

R851-R861.

McDonough, A. A. and D. Biemesderfer (2003). "Does membrane trafficking play a role in

regulating the sodium/hydrogen exchanger isoform 3 in the proximal tubule?" Curr

Opin Nephrol Hypertens 12(5): 533-41.

Meneton, P., X. Jeunemaitre, et al. (2005). "Links between dietary salt intake, renal salt

handling, blood pressure, and cardiovascular diseases." Physiol Rev 85(2):

679-715.

Nesher, M., M. Dvela, et al. (2009). "Physiological roles of endogenous ouabain in normal

rats." Am J Physiol Heart Circ Physiol 297(6): H2026-34.

Oweis, S., L. Wu, et al. (2006). "Cardiac glycoside downregulates NHE3 activity and

expression in LLC-PK1 cells." Am J Physiol Renal Physiol 290(5): F997-1008.

Schoner, W. and G. Scheiner-Bobis (2008). "Role of endogenous cardiotonic steroids in

sodium homeostasis." Nephrol Dial Transplant 23(9): 2723-9.

Schultheis, P. J., L. L. Clarke, et al. (1998). "Renal and intestinal absorptive defects in mice

lacking the NHE3 Na+/H+ exchanger." Nat Genet 19(3): 282-5.

Soleimani, M., B. A. Watts, 3rd, et al. (1998). "Effect of long-term hyperosmolality on the

Na+/H+ exchanger isoform NHE-3 in LLC-PK1 cells." Kidney Int 53(2): 423-31.

Stamler, J., G. Rose, et al. (1991). "Findings of the International Cooperative INTERSALT

Study." Hypertension 17(1 Suppl): I9-15.

Sweadner, K. J. (1989). "Isozymes of the Na+/K+-ATPase." Biochim Biophys Acta 988(2):

185-220.

77 Yang, L. E., A. B. Maunsbach, et al. (2004). "Differential traffic of proximal tubule Na+

transporters during hypertension or PTH: NHE3 to base of microvilli vs. NaPi2 to

endosomes." Am J Physiol Renal Physiol 287(5): F896-906.

Yates, N. A. and J. G. McDougall (1993). "Effects of direct renal arterial infusion of

bufalin and ouabain in conscious sheep." Br J Pharmacol 108(3): 627-30.

78 2.7 Figure Legends

Figure 2-1: Dose-dependent effects of ouabain (Oua) on Na/K-ATPase activity.

The HK-2, LLC-PK1 and AAC-19 cells were grown in 12-well plates with

Transwell membrane support to form monolayer. The Na/K-ATPase activity

(ouabain-sensitive 86Rb+ uptake) was assayed as described in Experimental Methods.

Data were shown as percentage of control, and each point is presented as mean ± S.E. of four sets of independent experiments. Curve fit analysis was performed by

GraphPad software.

Figure 2-2: Ouabain (Oua) inhibits H+-driven 22Na+ uptakes.

The HK-2, LLC-PK1 and AAC-19 cells were grown in 12-well plates to form

a monolayer. After treatment with ouabain (1h) and/or PP2 (1µM for 30min), 22Na+ was added and assayed for H+-driven 22Na+ uptake. To determine H+-driven Na+

+ uptake, cells were first acid loaded in Na -free buffer with 20 mM NH4Cl and then

assayed for 22Na+ uptake. 50 µM amiloride was used to inhibit amiloride-sensitive

NHE1 activity. Data are shown as mean ± S.E., percentage of control. n=4. ** p<0.01 compared to Control.

Figure 2-3: Ouabain (Oua) inhibits transcellular 22Na+ flux.

The HK-2, LLC-PK1 and AAC-19 cells were grown in 12-well plates with

Transwell membrane support to form a monolayer. The cells were treated with

ouabain (1h) and/or PP2 (1µM for 30min) in the basolateral or apical aspect. Active

79 transepithelial 22Na+ flux (apical to basolateral) was determined by counting radioactivity in the basolateral aspect at 1 h after 22Na+ addition. 50 µM amiloride

was added in the basolateral aspect to inhibit amiloride-sensitive NHE1 activity.

Data are shown as mean ± S.E., percentage of control. n=4. ** p<0.01 compared to

Control.

Figure 2-4: Ouabain (Oua) reduces cell surface expression of the α1 and NHE3.

The HK-2, LLC-PK1 and AAC-19 cells were treated with indicated

concentrations of ouabain (1h). Biotinylation of cell surface proteins was performed

to assess cell surface protein contents. Data are shown as mean ± S.E., percentage of control (Con). n=4. ** p<0.01 compared to control. Insert shows a representative western blot of four separate experiments. Immunoblotting with antibody against integrin-β1 served as loading controls (data not shown).

Figure 2-5: Ouabain (Oua) inhibits endocytic recycling of NHE3 in LLC-PK1 cells.

The experiments were performed as described in Experimental Methods. (A)

Determination of GSH-cleavage efficiency. Cell surface proteins were biotinylated and applied with or without GSH cleavage procedure. (B) Retrieval of total internalized biotinylated α1 and NHE3 after treatment with or without ouabain

(100nM, 1h) and GSH cleavage. (C) Assessment of the effect of ouabain on reinsertion. (D) The graph bars represented reinsertion of endocytosed α1 and NHE3,

80 the difference of total endocytosed α1 and NHE3 before and after resinsertion procedure. n=3. ** , p<0.01.

Figure 2-6: Ouabain (Oua) accumulates the Na/K-ATPase α1 subunit, but not

NHE3 in late endosome in LLC-PK1 cells.

LLC-PK1 cells were treated with or without ouabain (100nM for 1h), with or without pretreatment of chloroquine (Chlor, 0.2 mM, pretreated for 2h). Early endosome (EE) and late endosome (LE) fractions were isolated at the end of ouabain treatment. Equal amount of proteins (25 μg) was used to determine protein contents of the α1 and NHE3 by western blot analysis. n=4. ** p<0.01 compared to controls from EE and LE, respectively. # p<0.01, comparison of α1 subunit in LE fraction with or without chloroquine pretreatment. Insert shows a representative western blot of four separate experiments. Immunoblotting with antibodies against EEA-1 (for

EE fractionation) and Rab7 (for LE fractionation) served as loading controls, respectively (data not shown).

81 2.8 Tables and figures

Table 2.1: Ouabain causes a dose-dependent inhibition of transcellular 22Na+ flux, 22Na+ uptake, and surface α1 and NHE3. 22Na+ flux and 22Na+ uptake were measured as described in Experimental Methods. Surface α1 and NHE3 were determined by cell surface biotinylation. N=3 for each treatment. a, p<0.05 compared to controls; b, p<0.01 compared to controls.

HK-2 cells

Ouabain (nM) for 1h

0 1 5 10 50

22Na+ flux 100±4.5 102.1±3.8 81.2±5.2a 68.5±4.8b 61.8±5.9 b

22Na+ uptake 100±5.1 98.4±4.2 83.6±5.6a 76.6±6.2 b 67.8±5.3 b

Surface α1 100±6.1 96.5±6.7 84.2±6.2 a 63.5±7.1 b 59.7±7.4 b

Surface NHE3 100±4.5 99.2±6.1 86.2±6.8 a 67.3±7.1 b 69.1±7.9 b

LLC-PK1 cells

Ouabain (nM) for 1h

0 10 25 100 1,000

22Na+ flux 100±5.2 99.1±4.8 82.4±7.4 a 65.5±6.8 b 60.2±8.6 b

22Na+ uptake 100±5.8 95.7±6.7 88.3±8.1 a 64.8±7.2 b 58.5±7.8 b

Surface α1 100±7.9 97.3±8.1 85.6±7.2 a 65.2±7.7 b 67.6±8.4 b

Surface NHE3 100±6.5 98.6±8.8 83.2±7.4 a 72.7±6.1 b 69.3±7.6 b

82

AAC-19 cells

Ouabain (nM) for 1h

0 100 1,000 10,000 25,000

22Na+ flux 100±6.2 96.3±3.8 86.4±7.3 a 75.5±6.9 b 68.8±7.6 b

22Na+ uptake 100±7.2 97.8±7.1 85.1±8.5 a 71.6±8.1 b 65.2±9.6 b

Surface α1 100±8.2 96.5±7.6 87.4±8.1 a 75.2±7.9 b 67.4±9.1 b

Surface NHE3 100±9.1 94.7±8.7 84.8±9.3 a 69.5±8.8 b 61.8±9.9 b

83 Figure 2-1

84 Figure 2-2

85 Figure 2-3

86 Figure 2-4

87 Figure 2-5

88 Figure 2-6

89 Chapter 3 – Manuscript 2

Title:

Impairment of Na/K-ATPase Signaling in Renal Proximal Tubule Contributes to

Dahl Salt-Sensitive Hypertension

Authors:

Jiang Liu1, Yanling Yang1,3, Lijun Liu2, Zijian Xie2,1, Deepak Malhotra1, Bina Joe1, and

Joseph I. Shapiro1,2

From the Department of Medicine1, Physiology and Pharmacology2, University of Toledo

College of Medicine, Toledo, OH; and Institute of Biomedical Engineering3, Yanshan

University, China.

Running head : Na/K-ATPase signaling in salt sensitivity

Corresponding author:

Jiang Liu, Department of Medicine, University of Toledo College of Medicine, Health

Science Campus, 3000 Arlington Avenue, Toledo, Ohio 43614-2598.

Tel : 419-383-3923;

Email: [email protected]

Published in the Journal of Biological Chemistry. 2011, 286: 22806-22813.

90 3.1 Abstract

We have observed that, in renal proximal tubular cells, cardiotonic steroids such as ouabain in vitro signal through Na/K-ATPase which results in inhibition of transepithelial

22Na+ transport by redistributing Na/K-ATPase and NHE3. In the present study, we

investigate the role of Na/K-ATPase signaling in renal sodium excretion and blood

pressure regulation in vivo.

In Sprague Dawley rats, high salt diet activated c-Src and induced redistribution of

Na/K-ATPase and NHE3 in renal proximal tubules. In Dahl salt sensitive (S) and resistant

(R) rats given high dietary salt, we found different effects on blood pressure but, more

interestingly, different effects on renal salt handling. These differences could be explained by different signaling through the proximal tubular Na/K-ATPase. Specifically, in Dahl R

rats, high salt diet significantly stimulated phosphorylation of c-Src and ERK1/2, reduced

Na/K-ATPase activity and NHE3 activity, and caused redistribution of Na/K-ATPase and

NHE3. In contrast, these adaptations were either much less effective or not seen in the Dahl

S rats. To correct for the differences to high dietary salt, we also studied the primary culture

of renal proximal tubule isolated from Dahl S and R rats fed a low salt diet. In this system,

ouabain induced Na/K-ATPase/c-Src signaling and redistribution of Na/K-ATPase and

NHE3 in the Dahl R rats, but not in the Dahl S rats. Our data suggested that impairment of

Na/K-ATPase signaling and consequent regulation of Na/K-ATPase and NHE3 in renal proximal tubule may contribute to salt induced hypertension in the Dahl S rat.

91 3.2 Introduction

The Dahl salt resistant (R) and sensitive (S) strains were developed by selective

breeding of the outbred Sprague Dawley rat strain for resistance or susceptibility to the

hypertensive effects of high dietary sodium (Dahl, Heine et al. 1962). While the blood pressure (BP) response to salt-loading in Dahl R and S rats involves many regulatory factors (Rapp 1982), it has been proposed that renal proximal tubule (RPT) Na+ handling may be a critical determinant of the different BP responses in these strains (Dahl, Knudsen et al. 1969; Dahl, Heine et al. 1974).

Cardiotonic steroids (CTS) such as ouabain and marinobufagenin (MBG) appear to be involved in the regulation of BP and renal Na+ handling in vivo (Fedorova, Lakatta et al.

2000; Fedorova, Talan et al. 2002; Periyasamy, Liu et al. 2005) as well as the ion handling

of both primary cultures and proximal tubular cell lines in vitro (Blaustein and Hamlyn

1991; Yuan, Manunta et al. 1993; Liu, Periyasamy et al. 2002; Liu, Kesiry et al. 2004;

Periyasamy, Liu et al. 2005; Blaustein and Hamlyn 2010). Based on this background, we

performed the following studies to examine whether the proximal tubules of Dahl R and S

rats had different responses to CTS.

3.3 Materials and Methods

3.3.1 Animals:

Male Sprague Dawley rats (290-310 g body weight) were purchased from Charles

River. Male, age-matched Dahl R (SS/JrHsd) and S (SR/JrHsd) rats were bred and

maintained in-house and used at the age of 12-14 weeks. Rats were maintained with 12hr

92 dark/light cycle and had ad libitum access to the food (Teklad lab animal diets from Harlan

Laboratories) and water. All animal experimentation was conducted in accordance with the

National Institutes of Health Guide for the Care and Use of Laboratory Animals using

protocols approved by the University of Toledo Institutional Animal Use and Care

Committee.

3.3.2 Chemicals and Antibodies:

All chemicals, except otherwise mentioned, were obtained from Sigma-Aldrich (St.

Louis, MO). EZ-Link sulfo-NHS-ss-biotin and ImmunoPure immobilized

streptavidin-agarose beads were from Pierce Biotechnology (Rockford, IL). Antibodies

against Rab7, integrin-β1 and c-Src were from Santa Cruz Biotechnology (Santa Cruz,

CA). Monoclonal antibody against the Na/K-ATPase α1 subunit (clone α6F) was from the

Hybridoma Bank of the University of Iowa (Iowa City, IA). Monoclonal antibodies against

NHE3 (clone 4F5) and early endosome antigen-1 (EEA1) were from Millipore Chemicon

(Temecula, CA). Polyclonal anti-Src [pY418] phosphospecific antibody was from

Invitrogen (Camarillo, CA). Antibodies against phosphor-p44/42 ERK1/2 (Thr202/Tyr204)

and p44/42 were from Cell Signaling (Danvers, MA). Type 2 collagenase (activity of

323U/mg) was from Worthington Biochemical Corp (Lakewood, NJ). Sterile Percoll was

from GE Healthcare (Piscataway, NJ). Biomol Green was from BIOMOL Research

Laboratories (Plymouth Meeting, PA). Radioactive 22Na+ was from DuPont NEN Life

Science (Boston, MA).

3.3.3 Experimental groups and treatments:

93 For salt-loading studies in Sprague Dawley rats, the rats were randomly divided into two groups (10 rats per group) after adjustment to the new environment. The rats were then fed with 0.3% NaCl (as low salt diet) or 4% NaCl chow for 7 days. For

salt-loading studies in Dahl rats, age-matched, male R and S rats (total 48 rats) were given

either 0.3% NaCl (low salt diet, n=12 per strain) or 2% NaCl (n=12 per strain) diet for 7

days. Only 2% NaCl loading was used with the Dahl rats because of the profound effects

on BP and food consumption (inferred from urinary Na+ excretion data, see Table 2)

salt-loading has on the S animals.

3.3.4 Tail-cuff measurement of BP:

At day 0 and 7, BP was measured in conscious rats by the tail-cuff plethysmography with the aid of a computerized system (Amplifier model 229, Monitor model 31, Test

chamber Model 306; IITC Life Science). All rats were first trained for BP measurement

over a period of 2 days before day 0 in order to accustom them to the apparatus and restraint (Kennedy, Vetteth et al. 2006). After that, the actual BP (average of at least three

measurements) was recorded.

3.3.5 Isolation and primary culture of RPTs:

RPTs were isolated from the outer cortices as described by Vinay et al (Vinay,

Gougoux et al. 1981) with minor modifications. Briefly, harvested kidneys were

decapsulated and rinsed with ice-cold oxygenized PBS. Outer cortices were dissected,

minced, and digested in digesting solution (oxygenized DMEM with 1mg/ml collagenase

Type 2, 0.5% filtered BSA Fraction V) four times at 37 ºC, 15min each. Pooled tubular

94 segments were further separated with 42% Percoll gradient (pH7.4) and the RPT segments

were collected from the lowest two bands. The enrichment and purity of RPT isolation was

determined by measuring RPT brush-border membrane vehicles (BBMVs) marker enzyme

alkaline phosphatase activity (~10 fold increases) and by microscopic examination.

For in vitro primary culture, RPTs were isolated from R and S rats fed with low salt

diet. Isolated RPTs were cultured in collagen I-coated culture dishes in DMEM/F-12

medium (1:1 mixture, with 10% FBS and Insulin-transferrin-sodium selenite media

supplement, Sigma) until 90-100% confluency in a 5% CO2-humidified incubator. Before

ouabain treatment, cells were serum-starved (cultured in medium with 0.5% FBS) for

16-18 h.

3.3.6 Cell fractionation and biochemical studies:

Early endosome (EE, EEA-1- and Rab5-pisitive) and late endosome (LE,

Rab-7-positive) fractions were isolated by sucrose flotation centrifugation as previously

reported (Periyasamy, Liu et al. 2005; Cai 2008). The enrichment of EE and LE fractions

was assessed by the EE marker EEA1 and LE marker Rab7, respectively; Equal amounts of

total proteins from EE or LE fraction of each sample were precipitated with trichloroacetic

acid for subsequent Western blot. Surface biotinylation studies were conducted as

previously described (Liu, Kesiry et al. 2004; Cai 2008). Crude membrane isolation and

Na/K-ATPase enzymatic activity determinations (each performed in triplicate) were performed as previously described (Liu, Periyasamy et al. 2002).

3.3.7 RPT BBMVs Preparation and NHE3 activity assay (22Na+ uptake):

95 RPT BBMVs were prepared from isolated RPTs using the protocol described by

Murer’s Laboratory (Biber, Stieger et al. 2007). After homogenization, BBMVs were

2+ isolated with Mg precipitation method (final concentration of MgCl2 is 12 mM, 15 min

on ice) and differential centrifugation. Alkaline phosphotase activity was enriched ~10 fold

by this procedure. NHE3 activity (H+-driven 22Na+ uptake) in RPT BBMVs was

determined in triplicate using the filtration protocol described by Moe’s Laboratory (Bacic,

Kaissling et al. 2003).

3.3.8 Measurement of c-Src phosphorylation:

RPT whole cell lysates were prepared with Nonidet P-40 buffer containing 1%

Nonidet P-40, 0.25% sodium deoxycholate, 50 mM NaCl, 50 mM HEPES, 10% Glycerol

(pH 7.4), 1 mM sodium vanadate, 0.5 mM sodium fluoride, 1 mM Phenylmethanesulfonyl fluoride, and protease inhibitor cocktail for general use (Sigma). After clarification, 300μg of total protein was immunoprecipitated with antibody against c-Src and protein G-agrose beads (Millipore), and then eluted with 2x Laemmli buffer. After immunoblotting for phospho-c-Src (p-Src), the same membrane was stripped and immunoblotted for total c-Src (t-Src). The activation of c-Src was expressed as ratio of p-Src/ t-Src with both measurements normalized to 1.0 for the control samples.

3.3.9 Western blotting:

For Western blot analysis, equal amounts of total protein were resolved by 10%

SDS-PAGE and immunoblotted with antibodies against interested proteins. The same membrane was also immunoblotted with antibodies against integrin-β1 (for surface

96 biotinylation) or EEA-1 (for EE fractionation) to serve as loading controls, respectively.

Signal detection was performed with an enhanced chemiluminescence super signal kit

(Pierce, Rockford, IL). Multiple exposures were analyzed to assure that the signals were

within the linear range of the film. The signal density was determined using Molecular

Analyst software (Bio-Rad, Hercules, CA).

3.3.10 Statistical analysis:

Data were tested for normality (all data passed) and then subjected to parametric

analysis. When more than two groups were compared, one-way ANOVA was performed

prior to comparison of individual groups, and the post-hoc t-tests were adjusted for

multiple comparisons using Bonferroni’s correction. Statistical significance was reported

at the P < 0.05 and P < 0.01 levels. SPSS software was used for all analysis. Values are given as mean±SEM (Wallenstein, Zucker et al. 1980).

3.4 Results

3.4.1 Effect of high salt diet on RPT Na/K-ATPase and NHE3 in Sprague Dawley

rats:

As shown in Table 1, the high salt diet significantly increased absolute urinary Na+ excretion compared with control rats. The high salt diet also had a small effect on systolic

BP. The high salt diet reduced the Na/K-ATPase activity in RPT crude plasma membrane fractions and NHE3 activity in RPT BBMVs (Table 1). In addition, the high salt diet not only decreased RPT surface contents of both transporters (Fig. 3-1a), but also caused both

97 transporters in RPTs to accumulate in the EE fraction (Fig. 3-1b). The inhibition of the

Na/k-ATPase activity and NHE3 activity was well correlated with reduced protein content

of the α1 subunit and NHE3 on the cell surface. Furthermore, the high salt diet stimulated

c-Src phosphorylation in RPT whole cell lysates (Fig.3-1c).

RPT primary cultures isolated from control rats fed with low salt diet were treated

with ouabain or vehicle. Ouabain treatment (10 µmol/L for 1 h) accumulated the α1 subunit

and NHE3 in the EE fractions, reduced the surface protein content of these two ion

transporters (Fig. 3-2a), and caused an inhibition of both Na/K-ATPase activity (in

µmol/mg protein/hour, control 58.9±3.6 vs. ouabain 48.1±3.1, n=4, p<0.01 compared to

control) and NHE3 activities by measuring H+-driven 22Na+ uptake (in relative value,

control 100±4.5 vs. ouabain-treated 76.5±6.8, n=4, p<0.01 compared to control). Moreover,

ouabain (10 or 25 µmol/L for 5 min) stimulated c-Src phosphorylation (Fig. 3-2b).

3.4.2 Effect of high salt diet on RPT Na/K-ATPase and NHE3 from Dahl R and S

rats:

Male age-matched Dahl R and S rats were given a low salt (0.3% NaCl, n=12 per

strain) or high salt (2% NaCl, n=12 per strain) diet for 7 days. As briefly mentioned earlier,

we used only a 2% high salt diet as the Dahl S (but not R) rats showed profound increases

in blood pressure as well as very substantial decreases in food consumed as evidenced by

the relative decrease in UNa+V compared with the R rats (Table 2). There were no

significant differences amongst the groups in the plasma concentrations of Na+ and K+ at the end of the experiments. These and additional data are summarized in Table 2.

98 In the isolated RPTs from Dahl R and S rats fed a low salt diet, there were no differences in the expression of Na/K-ATPase α1 subunit and NHE3 or Na/K-ATPase and

NHE3 activities. High salt diet has no significant effect on total expression of

Na/K-ATPase α1 and NHE3 in isolated RPTs from the R and S rats (data not shown).

However, the high salt diet significantly decreased both Na/K-ATPase and NHE3 activities in the R but not the S rats (Fig.3-3).

In the isolated RPTs from the R rats, the high salt diet not only decreased the RPT surface content of both Na/K-ATPase α1 and NHE3 (Fig.3-4a) but also caused the accumulation of both transporters in the EE fraction (Fig.3-4b). Interestingly, the high salt diet did not cause redistribution of the α1 subunit and NHE3 in the S rat RPTs (Fig. 3-4a and 4b). While the high salt diet caused the α1 subunit to also accumulate in the LE fractions in the R rats (relative to control 100±3.5, high salt diet 246.5±7.6, n=3, p<0.01), the NHE3 did not accumulate in the LE fraction (Fig.3-4c). This suggests that the NHE3 which moves to the EE fraction remains available for recycling, but in any case doesn’t accumulate in the LE fraction.

In the RPTs isolated from the R but not the S rats, high salt diet stimulated activation of c-Src (Fig. 3-5a) and ERK1/2 (Fig. 3-5b). In the same immunoprecipitation studies shown in Fig. 3-5a, high salt diet also significantly enhanced association between c-Src and

Na/K-ATPase α1 (relative to the low salt control diet 100±3.7, high salt diet 184.6±9.3, n=3, p<0.01) in the R rats, but not in the S rats.

3.4.3 Effects of Ouabain on RPT primary cultures derived from Dahl R and S rats:

99 RPTs were isolated from age-matched male R and S rats (20 R rats and 20 S rats) fed

with a low salt diet (0.3% NaCl), and cultured to reach confluence. As observed in

salt-loading experiment with intact animals, ouabain treatment also caused differential

regulations in the RPT primary culture derived from the R and S rats. While ouabain

demonstrated no regulatory effect in the RPTs derived from the S rats, ouabain had

dramatic consequences in the RPTs derived from the R rats. Specifically, ouabain treatment (10 µmol/L for 1 h) caused the accumulation of the α1 subunit and NHE3 in the

EE fractions (Fig. 3-6a); reduced the surface protein content of these two ion transporters

(Fig. 3-6b); and inhibited both the membrane Na/K-ATPase and NHE3 activities in RPT

crude membrane fraction and BBMVs, respectively (Table 3). Moreover, ouabain

treatment (1 and 10 µmol/L for 5 min) stimulated phosphorylation of c-Src and ERK1/2

(Fig. 3-7a and 7b). In the same immunoprecipitation studies shown in Fig. 3-7a, ouabain

also enhanced the association between c-Src and Na/K-ATPase α1 (in values relative to

control 100±4.2, ouabain 1 µmol/L 167.6±8.2 and ouabain 10 µmol/L 192.8±6.3, n=4,

both p<0.01 compared to control) in the R rats. No significant association between c-Src

and Na/K-ATPase α1 was observed in the S rats in response to either dose of ouabain.

3.5 Discussion

The significance of endogenous CTS functioning as natriuretic hormones by

inhibiting the renal Na/K-ATPase has been a subject of debate for some time (Bagrov,

Shapiro et al. 2009). While volume expansion is considered to be the main cause of the

100 salt-sensitive hypertension in the S rats (Rapp 1982; Roman and Osborn 1987), accumulating evidence, some from our laboratories, suggest that endogenous CTS regulate renal Na+ handling and BP through the Na/K-ATPase signaling (Fedorova, Lakatta et al.

2000; Fedorova, Talan et al. 2002; Dostanic, Paul et al. 2005; Zhang, Lee et al. 2005;

Loreaux, Kaul et al. 2008; Nesher, Dvela et al. 2009; Fedorova OV 2010; Liu and Xie

2010). A role for endogenous CTS in salt sensitive hypertension has also been suggested by a number of authors (Schoner and Scheiner-Bobis 2008; Blaustein and Hamlyn 2010;

Jaitovich and Bertorello 2010; Ritz 2010). On this background, the Dahl R and S rats have been extensively studied for insight into the pathogenesis of salt sensitive hypertension.

Fedorova and coworkers have established that increases in circulating MBG are, in fact, greater during acute and chronic salt-loading in Dahl S as compared with R rats (Fedorova,

Lakatta et al. 2000; Fedorova, Talan et al. 2002).

In the current study, we observed that the outbred Sprague Dawley rats developed redistribution of the Na/K-ATPase in RPTs in response to a shift to a high salt diet. As we have previously observed (Periyasamy, Liu et al. 2005), this redistribution of the

Na/K-ATPase appears to depend on increases in the production of the CTS, MBG. In addition to what we had previously seen, we also noted what appeared to be a coordinated redistribution of the apical NHE3 into the EE but not LE fraction, a phenomenon which we have documented to be a consequence of Na/K-ATPase-c-Src signaling in LLC-PK1 cells

(Cai 2008). Interestingly, works from the laboratory of McDonough using immunofluorescence have shown that a high salt diet causes the redistribution of NHE3 to a deeper portion of microvilli whereas this high salt diet causes the sodium-phosphate

101 cotransporter to move completely into cells (Yang, Meng et al. 2008). We believe that our

data with the NHE3 are consistent with this report.

We further demonstrated that CTS signaling also appeared to occur in a similar fashion in Dahl R but not S rats. Specifically, we saw that high salt diet induced redistribution of the Na/K-ATPase and NHE3 along with evidence for c-Src and ERK1/2

activation in RPTs isolated from the Dahl R but not S rats. Impaired CTS signaling in the

RPTs of the S rats was further implicated by the failure of these RPTs isolated from the S

rats on a low salt diet to show ouabain signaling in vitro, which was readily apparent in

parallel studies performed in the RPTs isolated from the R rats. As the S rats developed

profound hypertension when dietary Na+ is increased to 2% whereas the R rat

demonstrated little change in BP with this dietary manipulation, these data implicated the

CTS-Na/K-ATPase-c-Src signal cascade in the control of BP in these animals.

Furthermore, since the S rats develop greater increases in the production of as well as

circulating levels of CTS following increases in dietary Na+ (Fedorova, Lakatta et al. 2000;

Fedorova, Talan et al. 2002), diminished sensitivity of the signaling cascade to CTS would

appear to be involved. However, as the α1 subunit sequence of the Na/K-ATPase does not

differ between Dahl S and R rats (Mokry and Cuppen 2008), one must implicate the

environment of the signaling Na/K-ATPase and/or the interactions between the

Na/K-ATPase and its signaling partners (Pierre and Xie 2006).

We believe that these data are interesting for several reasons. First, they implicate

CTS in the pathogenesis of salt sensitive hypertension by a renal mechanism, namely the

failure of the Dahl S to demonstrate the normal response to CTS seen with the Dahl R and

the outbred Sprague Dawley rats. This may give pause to implementing the therapeutic

102 strategy of antagonizing CTS-Na/K-ATPase signaling in vascular tissue which has been proposed by pioneering workers in this field (Yuan, Manunta et al. 1993; Blaustein and

Hamlyn 2010). Second, these data suggest that CTS may induce increases in renal sodium excretion by multiple mechanisms. Specifically, these agents may increase vascular resistance as others have demonstrated (Bagrov and Fedorova 2005; Fedorova, Kolodkin et al. 2005; Kashkin, Zvartau et al. 2007) while also working through the renal mechanism

discussed above. Although other hormonal systems no doubt interact with both of these

pathways (Fedorova, Agalakova et al. 2006), it does appear that the former mechanism is

much more intact in the Dahl S rats suggesting that the molecular mechanisms of CTS

signaling might be different in these different tissues. Whether this is explained by

Na/K-ATPase isoform differences, differences in specific CTS and/or mechanistic

differences in the way that CTS signal in vascular as compared to renal tissues is still quite

unclear. On this latter point, we might speculate that the classic ionic signal mechanism

involving actual pump inhibition might dominate in vascular tissue as proposed by

Blaustein and Hamlyn (Blaustein and Hamlyn 2010) whereas the CTS-c-Src signaling

might be more important in the RPTs. Of course, further work must be done to address this

complex area. Last, these data might possibly provide the basis for developing a tool for

clinical practice. If the sensitivity of the Na/K-ATPase-c-Src signaling pathway to CTS is

something that is impaired either on a genetic or acquired basis, it might be possible to use

this phenomenon to develop a biomarker for salt sensitive hypertension.

103 Acknowledgments:

Portions of this work were supported by grants from National Institutes of Health

(GM78565, HL36573, HL020176 and HL076709) and American Heart Association Ohio

Valley Affiliate.

104 3.6 References

Bacic, D., B. Kaissling, et al. (2003). "Dopamine acutely decreases apical membrane Na/H

exchanger NHE3 protein in mouse renal proximal tubule." Kidney Int 64(6): 2133-41.

Bagrov, A. Y. and O. V. Fedorova (2005). "Cardenolide and bufadienolide ligands of the

sodium pump. How they work together in NaCl sensitive hypertension." Front Biosci 10:

2250-6.

Bagrov, A. Y., J. I. Shapiro, et al. (2009). "Endogenous cardiotonic steroids: physiology,

pharmacology, and novel therapeutic targets." Pharmacol Rev 61(1): 9-38.

Biber, J., B. Stieger, et al. (2007). "Isolation of renal proximal tubular brush-border

membranes." Nat Protoc 2(6): 1356-9.

Blaustein, M. P. and J. M. Hamlyn (1991). "Pathogenesis of essential hypertension. A link

between dietary salt and high blood pressure." Hypertension 18(5 Suppl): III184-95.

Blaustein, M. P. and J. M. Hamlyn (2010). "Signaling mechanisms that link salt retention

to hypertension: Endogenous ouabain, the Na(+) pump, the Na(+)/Ca(2+) exchanger and

TRPC proteins." Biochim Biophys Acta 1802(12): 1219-29.

Cai, H., Wu, L., Qu, W., Malhotra, D., Xie, Z., Shapiro, JI, and Liu, J. (2008). "Regulation

of Apical NHE3 Trafficking by Ouabain-Induced Activation of Basolateral Na/K-ATPase

Receptor Complex." Am J Physiol Cell Physiol 294(2): C555-63.

Dahl, L. K., M. Heine, et al. (1962). "Role of genetic factors in susceptibility to

experimental hypertension due to chronic excess salt ingestion." Nature 194: 480-2.

105 Dahl, L. K., M. Heine, et al. (1974). "Genetic influence of the kidneys on blood pressure.

Evidence from chronic renal homografts in rats with opposite predispositions to

hypertension." Circ Res 40(4): 94-101.

Dahl, L. K., K. D. Knudsen, et al. (1969). "Humoral transmission of hypertension:

evidence from parabiosis." Circ Res 24(5): Suppl:21-33.

Dostanic, I., R. J. Paul, et al. (2005). "The alpha2-isoform of Na-K-ATPase mediates

ouabain-induced hypertension in mice and increased vascular contractility in vitro." Am J

Physiol Heart Circ Physiol 288(2): H477-85.

Fedorova, O. V., N. I. Agalakova, et al. (2006). "ANP differentially modulates

marinobufagenin-induced sodium pump inhibition in kidney and aorta." Hypertension

48(6): 1160-8.

Fedorova, O. V., N. I. Kolodkin, et al. (2005). "Antibody to marinobufagenin lowers blood pressure in pregnant rats on a high NaCl intake." J Hypertens 23(4): 835-42.

Fedorova, O. V., E. G. Lakatta, et al. (2000). "Endogenous Na,K pump ligands are

differentially regulated during acute NaCl loading of Dahl rats." Circulation 102(24):

3009-14.

Fedorova OV, S. J., Bagrov AY. (2010). "Endogenous cardiotonic steroids and salt-sensitive hypertension." Biochimica et Biophysica Acta (BBA) - Molecular Basis of

Disease doi:10.1016/j.bbadis.2010.03.011 (Epub ahead).

Fedorova, O. V., M. I. Talan, et al. (2002). "Endogenous ligand of alpha(1) sodium pump,

marinobufagenin, is a novel mediator of sodium chloride--dependent hypertension."

Circulation 105(9): 1122-7.

106 Jaitovich, A. and A. M. Bertorello (2010). "Salt, Na+,K+-ATPase and hypertension." Life

Sci 86(3-4): 73-8.

Kashkin, V. A., E. E. Zvartau, et al. (2007). "Endogenous bufadienolide mediates pressor response to ethanol withdrawal in rats." Eur Neuropsychopharmacol.

Kennedy, D. J., S. Vetteth, et al. (2006). "Central role for the cardiotonic steroid marinobufagenin in the pathogenesis of experimental uremic cardiomyopathy."

Hypertension 47(3): 488-95.

Liu, J., R. Kesiry, et al. (2004). "Ouabain induces endocytosis of plasmalemmal

Na/K-ATPase in LLC-PK1 cells by a clathrin-dependent mechanism." Kidney Int 66(1):

227-41.

Liu, J., S. M. Periyasamy, et al. (2002). "Effects of cardiac glycosides on sodium pump expression and function in LLC-PK1 and MDCK cells." Kidney Int 62(6): 2118-25.

Liu, J. and Z. J. Xie (2010). "The sodium pump and cardiotonic steroids-induced signal transduction protein kinases and calcium-signaling microdomain in regulation of transporter trafficking." Biochim Biophys Acta: (10.1016/j.bbadis.2010.01.013) [Epub

ahead of print].

Loreaux, E. L., B. Kaul, et al. (2008). "Ouabain-Sensitive alpha1 Na,K-ATPase enhances

natriuretic response to saline load." J Am Soc Nephrol 19(10): 1947-54.

Mokry, M. and E. Cuppen (2008). "The Atp1a1 gene from inbred Dahl salt sensitive rats

does not contain the A1079T missense transversion." Hypertension 51(4): 922-7.

Nesher, M., M. Dvela, et al. (2009). "Physiological roles of endogenous ouabain in normal rats." Am J Physiol Heart Circ Physiol 297(6): H2026-34.

107 Periyasamy, S. M., J. Liu, et al. (2005). "Salt loading induces redistribution of the

plasmalemmal Na/K-ATPase in proximal tubule cells." Kidney Int 67(5): 1868-77.

Pierre, S. V. and Z. Xie (2006). "The Na,K-ATPase receptor complex: its organization and

membership." Cell Biochem Biophys 46(3): 303-16.

Rapp, J. P. (1982). "Dahl salt-susceptible and salt-resistant rats. A review." Hypertension

4(6): 753-63.

Ritz, E. (2010). "Salt and hypertension." Nephrology (Carlton) 15 Suppl 2: 49-52.

Roman, R. J. and J. L. Osborn (1987). "Renal function and sodium balance in conscious

Dahl S and R rats." Am J Physiol 252(5 Pt 2): R833-41.

Schoner, W. and G. Scheiner-Bobis (2008). "Role of endogenous cardiotonic steroids in

sodium homeostasis." Nephrol Dial Transplant 23(9): 2723-9.

Vinay, P., A. Gougoux, et al. (1981). "Isolation of a pure suspension of rat proximal

tubules." Am J Physiol 241(4): F403-11.

Wallenstein, S., C. L. Zucker, et al. (1980). "Some statistical methods useful in circulation

research." Circ Res 47(1): 1-9.

Yang, J. H., X. X. Meng, et al. (2008). "Acute myocardial ischemia up-regulates substance

P in the retina of rats." Neurosci Lett 443(3): 218-22.

Yuan, C., P. Manunta, et al. (1993). "Role of ouabain-like factors in hypertension: effects

of ouabain and certain endogenous ouabain-like factors in hypertension." J Cardiovasc

Pharmacol 22 Suppl 2: S10-2.

Zhang, J., M. Y. Lee, et al. (2005). "Sodium pump alpha2 subunits control myogenic tone

and blood pressure in mice." J Physiol 569(Pt 1): 243-56.

108 3.7 Figure Legends

Figure 3-1: In Sprague Dawley rats, 7-day high salt diet (HS) activated c-Src and stimulated redistribution of RPT Na/K-ATPase α1 subunit and NHE3.

HS reduced RPT cell surface contents of the α1 subunit and NHE3, n=5 (a), accumulated the α1 subunit and NHE3 in RPT EE fractions, n=5 (b), and stimulated c-Src phosphorylation in RPT whole cell lysates, n=4 (c). ** p<0.01 compared to control. RPT isolation, surface biotinylation and EE fraction isolation from control and treated rats were conducted in pairs concurrently. Immunoblotting of integrin-β1 and EEA-1 was served as loading control for surface biotinylation and EE fraction, respectively.

Figure 3-2:

In RPT primary culture derived from Sprague Dawley rats, ouabain (Oua) not only redistributed the Na/K-ATPase α1 subunit and NHE3, n=5 (a), but it also stimulated c-Src phosphorylation within 5 minute in RPTs, n=4 (b). ** p<0.01 compared to control. The experiments in (a) were performed as in Figure 2-1, with integrin-β1 and EEA-1 as loading control.

Figure 3-3:

In the R rats, high salt diet (HS) inhibited both the Na/K-ATPase activity (a) and

NHE3 activity (b). Crude heavy plasma membrane fractions and RPT BBMVs were prepared from isolated RPTs at the end of experiments. The Na/K-ATPase enzymatic

109 activity (µmol/mg protein/hour) and relative NHE3 activity were calibrated by protein

content. n=6. ** p<0.01 compared to control.

Figure 3-4: In Dahl rats, high salt diet (HS) redistributed the Na/K-ATPase α1 and

NHE3 in RPTs isolated from the R rats, but not in the S rats.

Male, age-matched Dahl R and S rats were paired for salt-loading. Cell surface α1

subunit and NHE3 were determined by surface biotinylation (a) with integrin-β1 blotting

as loading control, and internalized α1 subunit and NHE3 were determined by the protein

content in EE fractions (b) with EEA-1 blotting as loading control. n=6. ** p<0.01

compared to control. In LE fractions, high salt diet accumulated α1 subunit only in the R

rats, but NHE3 protein content was not significantly affected by high salt diet in both R and

S rats (c). Rab7 blotting was served as loading control.

Figure 3-5: High salt diet (HS) activated c-Src and ERK1/2 and enhanced association

between c-Src and the α1 subunit in RPTs of the R rats, but not the S rats.

(a) HS stimulated c-Src phosphorylation and its association with the α1 subunit. (b)

HS activated ERK1/2. Values are expressed relative to a control value of 100. n=4. **, p

<0.01 compared to control.

Figure 3-6: Ouabain (Oua, 10µM for 1 hour) redistributed the Na/K-ATPase α1

subunit and NHE3 in RPT primary culture of the R rats, but not the S rats.

RPTs were isolated from the R and S rats fed with low salt (0.3% NaCl) diet. Cell surface NHE3 and α1 subunit were determined by surface biotinylation (a), and

110 internalized NHE3 and α1 subunit were determined by the protein content in EE fractions

(b). n=4. ** p<0.01 compared to control. Immunoblotting of integrin-β1 and EEA-1 was served as loading control for surface biotinylation and EE fraction, respectively.

Figure 3-7: Ouabain (Oua, 1 and 10µM for 5 min) activated c-Src and ERK1/2 and enhanced association between c-Src and the α1 subunit in RPTs of the R rats, but not the S rats.

(a) ouabain stimulated c-Src phosphorylation and its association with the α1 subunit.

(b) ouabain activated ERK1/2. Values are expressed relative to a control value of 100. n=4.

**, p<0.01.

111 3.8 Tables and figures

Table 3.1: The effect of high salt diet (4% NaCl for 7 days) on renal sodium excretion

of Sprague Dawley rats.

Absolute Na+ excretion (UNa+V) was calculated as urinary Na+ concentration x

24-h urine volume. The Na/K-ATPase activity was measured from isolated

RPTs and expressed as µmol/mg protein/hour from four independent

experiments (each performed in triplicate). NHE3 activity (22Na+ uptake) in

BBMVs was measured from isolated RPTs and expressed as relative values (%,

control was normalized as 100%) from four independent experiments (each

performed in triplicate). ** p<0.01 compared to control. n=rat number (for

UNa+V and BP) or experiment number (for Na/K-ATPase and NHE3 activity).

UNa+V (mEq/24h) BP (mmHg) Na/K-ATPase activity NHE3 activity (%)

Control 2.7 ± 0.4 (n=10) 102.6±2.5 (n=10) 56.4±4.2 (n=4) 100±4.8 (n=4)

High salt diet 18.1 ± 1.7(n=10)** 115.3±3.2(n=10)** 40.2±6.8 (n=4) ** 67.6±10.5 (n=4) **

112 Table 3.2: High salt diet (HS, 2% NaCl, 7 days) caused differential regulation

between the R and S rats. Absolute Na+ excretion (UNa+V) was calculated as

Urine Na+ x 24-h urine volume. Fractional Na+ excretion (FENa+) was

calculated as 100 x (Urine Na+ x Plasma creatinine)/(Plasma Na+ x Urine

creatinine). Creatinine clearance was calculated as (Urine creatinine x 24-h

urine volume)/(Plasma creatinine x 24 x 60min). ** p<0.01 compared to

control LS diet of the same strain. # p<0.01 the R rats with HS compared to the

S rats with HS. n=rat number, per treatment, per group.

Strain LS HS

BP R 133±2.2 (n=12) 134±2.6 # (n=12)

S 146±3.1 (n=12) 169±5.6** (n=12)

Plasma Na+ R 145±2.8 (n=12) 146±1.4 (n=12)

S 146±2.1 (n=12) 147±3.6 (n=12)

Plasma K+ R 8.7±1.8 (n=12) 8.6±1.3 (n=12)

S 8.8±1.4 (n=12) 8.7±1.5 (n=12)

UNa+V (mEq/24h) R 2.9±0.6(n=8) 20.1±1.6**,#(n=8)

S 2.7±0.25(n=8) 6.5±1.0**(n=8)

FENa+ R 0.073±0.008(n=8) 0.287±0.064**,#(n=8)

S 0.071±0.004(n=8) 0.132±0.021**(n=8)

Creatinine clearance R 1.99±0.14(n=8) 2.66±0.38(n=8)

(ml/min) S 2.06±0.32(n=8) 2.67±0.64(n=8)

113 Table 3.3: The effect of ouabain on the Na/K-ATPase activity and NHE3 activity in

RPT primary cultures derived from the R and S rats. The Na/K-ATPase

activity in crude heavy membrane fractions was expressed as µmol/mg

protein/hour from four independent experiments (each performed in triplicate).

NHE3 activity (22Na+ uptake) in BBMVs was expressed as relative values (%

of control as 100%) from four independent experiments (each performed in

triplicate). ** p<0.01 compared to control.

Strain Control Ouabain (10µM, 1h)

Na/K-ATPase activity R 61.2±3.6 (n=4) 46.8±3.5** (n=4)

S 60.8±4.1 (n=4) 57.9±4.5 (n=4)

NHE3 activity (%) R 100±5.2 (n=4) 71.2±5.5 **(n=4)

S 100±6.1 (n=4) 96.4±6.5 (n=4)

114 Figure 3-1

a. b.

c.

115 Figure 3-2 a.

b.

116 Figure 3-3 a.

75 LS HS

50 **

25 Enzymatyic activity (uMol/mg protein/hour) (uMol/mg

0 R rats S rats

b.

125 LS HS

100 uptake, %) + 75 ** Na 22

50

25 NHE3 activity ( 0 R rats S rats

117 Figure 3-4 a. b.

c.

118 Figur 3-5 a.

b.

119 Figure 3-6 a.

b.

120 Figure 3-7 a.

b.

121 Chapter 4 – Manuscript 3

Title:

Redox Modulation of the Na/K-ATPase Signaling and Renal Proximal Tubular

Sodium Handling

Authors:

Yanling Yan1,3, Vinai Katragadda1, Steven Haller1, Anna P. Shapiro1, Joe Xie1, Chiamaka

Mbaso1, Deepak Malhotra1, Zi-jian Xie2,1, Joseph I. Shapiro1,2, and Jiang Liu1

1Departments of Medicine and Physiology, University of Toledo College of

Medicine, Toledo, OH; and 3Institute of Biomedical Engineering, Yanshan

University, China.

In preparation.

122 4.1 Abstract

We have shown that cardiotonic steroids (CTS) signaling through the Na/K-

ATPase regulate renal proximal tubule (RPT) sodium reabsorption and impairment of the Na/K-ATPase signaling contributes to experimental Dahl salt-sensitive hypertension. Here we report that reactive oxygen species (ROS) is critical in modulation of ouabain-induced Na/K-ATPase signaling and RPT ion transport.

Antioxidant NAC (N-Acetyl-L-cysteine) prevented ouabain-stimulated Na/K-

ATPase signaling cascade, protein carbonylation, redistribution of the Na/K-

ATPase and NHE3, and inhibition of transepithelial 22Na+ transport. However, a basal physiological range of ROS is necessary for initiation and amplification of ouabain-Na/K-ATPase signaling and redistribution of Na/K-ATPase and NHE3.

Furthermore, both ouabain and glucose oxidase caused direct carbonylation of two amino acid residues in the actuator (A) domain of the Na/K-ATPase α1 subunit.

123 4.2 Introduction

Excessive dietary salt intake significantly contributes to the development of hypertension and tends to be more pronounced in typical salt-sensitive patients

(Calhoun, Jones et al. 2008). Renal proximal tubule (RPT) sodium handling, which

claims over 60% reabsorption of filtered sodium, is an independent determinant in

the pathogenesis of salt-sensitive hypertension. Accumulating data indicate that

CTS (also known as endogenous digitalis-like substances) signaling through the

Na/K-ATPase regulates RPT sodium handling and salt sensitivity (reviewed in

(Schoner and Scheiner-Bobis 2008; Fedorova, Shapiro et al. 2010; Liu and Xie

2010)).

It is well documented that a high salt diet stimulates an increase in

endogenous CTS release and ROS generation. Our recent observations indicate that

RPT sodium handling via ligand-mediated Na/K-ATPase/c-Src signaling

counterbalances the sodium retention mediated increases in blood pressure, such as

that seen in salt-sensitive hypertension (Liu, Kesiry et al. 2004; Liu, Liang et al.

2005; Periyasamy, Liu et al. 2005; Oweis, Wu et al. 2006; Cai, Wu et al. 2008; Liu,

Yan et al. 2011). Ouabain, a ligand of the Na/K-ATPase, activates the Na/K-

ATPase/c-Src signaling pathway and subsequently redistributes basolateral Na/K-

ATPase and apical sodium/hydrogen exchanger isoform 3 (NHE3) in RPTs, leading

to reduced RPT sodium reabsorption and increased urinary sodium excretion. We

have also demonstrated that impairment of the RPT Na/K-ATPase/c-Src signaling

contributes to experimental Dahl salt-sensitive hypertension (Liu, Yan et al. 2011).

Since it failed to confirm the possible difference of the Na/K-ATPase α1 gene

124 (Atp1a1) coding (Mokry and Cuppen 2008) and ouabain sensitivity of the Na/K-

ATPase (Nishi, Bertorello et al. 1993) between the Dahl salt-resistant (R) and salt-

sensitive (S) rats (Jr strains), other factor(s) must be present to prevent activation of

the Na/K-ATPase/c-Src signaling in the S rats. Normal physiological ROS signaling

is critical in systematic and cellular functions, and excessive oxidative stress

contributes to the development and maintenance of hypertension in animal models in which, the RPT sodium and fluids reabsorption through the Na/K-ATPase and

NHE3 activities are dysregulated (Zhang, Imam et al. 2002; Panico, Luo et al. 2009;

Banday and Lokhandwala 2011). Amongst ROS-induces many kinds of post-

translational modifications, protein direct carbonylation, which are mostly

happened in lysine, arginine, threonine, and proline residues, is widely used as a

biomarker of oxidative stress (Dalle-Donne, Rossi et al. 2003; Stadtman and Levine

2003). We present here that ouabain-stimulated Na/K-ATPase signaling and

subsequent redistribution are redox-sensitive, and ouabain induces direct

carbonylation of proline/threonine residues in the actuator (A) domain of the Na/K-

ATPase α1 subunit. The data indicate that a proper redox modulation of RPT Na/K-

ATPase signaling is critical in renal sodium handling and might play an important

role in counteracting volume expansion mediated high blood pressure.

4.3 Materials and Methods

4.3.1 Chemicals and Antibodies

125 All chemicals, except otherwise mentioned, were obtained from Sigma-

Aldrich (St. Louis, MO). EZ-Link sulfo-NHS-ss-biotin and ImmunoPure immobilized streptavidin-agarose beads were obtained from Pierce Biotechnology

(Rockford, IL). Monoclonal antibody against the Na/K-ATPase α1 subunit (clone

α6F) was from the Developmental Studies Hybridoma Bank at the University of

Iowa (Iowa City, IA). Monoclonal antibody against early endosome antigen-1

(EEA1) and OxyBlot Protein Oxidation Detection Kit were from Millipore

Chemicon (Temecula, CA). Polyclonal anti-Src [pY418] phosphospecific antibody was from Invitrogen (Camarillo, CA). Antibodies against phosphor-p44/42 ERK1/2

(Thr202/Tyr204) and p44/42 were from Cell Signaling Technology (Danvers, MA).

DNPH (2,4-Dinitrophenylhydrazine) and antibody against DNP was from Sigma.

DMPO (5,5-Dimethyl-1-Pyrroline-N-Oxide) and polyclonal anti-DMPO antiserum were from Cayman Chemicals (Ann Arbor, MI). Radioactive 22Na+ was from

DuPont NEN Life Science (Boston, MA).

4.3.2 Cell cultures

Porcine renal proximal tubule LLC-PK1 cells, mice SYF and SYF+c-Src cells were obtained from the American Type Culture Collection (Manassas, VA) and cultured with DMEM (Dulbecco's modified Eagle's medium with 10% FBS,

100 U/ml penicillin, and 100 µg/ml streptomycin) in a 5% CO2-humidified incubator. Culture medium was changed daily until confluence. LLC-PK1 cells were serum-starved for 16-18 h before treatment. In assays for active transcellular

22Na+ flux, cells were grown on Transwell membrane support to form monolayer,

126 and then treated with ouabain either in basolateral or apical compartment. For SYF

and SYF+c-Src cells, medium was changed daily until the cells reached 80-90%

confluence, at which time the medium was changed to DMEM with 1% FBS for at

least 12 hours before experiments.

4.3.3 Isolation of early endosome (EE) fraction

EE (EEA-1- and Rab5-pisitive) fraction was isolated based on a flotation

gradient using the technique of Gorvel (Gorvel, Chavrier et al. 1991) and identified

as we previously described (Liu, Kesiry et al. 2004; Cai, Wu et al. 2008). The enrichment of EE fraction was assessed by the EE marker EEA-1. Equal amounts of

total proteins from EE fraction of each sample were precipitated with trichloroacetic

acid for subsequent Western blot.

4.3.4 Active transepithelial 22Na+ flux assa

LLC-PK1 cells were cultured on Transwell membrane support (Costar

Transwell culture filter inserts, filter pore size: 0.4µM, Costar, Cambridge, MA ) to

form monolayers. Active transepithelial 22Na+ flux (from apical to basolateral aspect)

was determined by counting radioactivity in the basolateral aspect at 1 hour after

addition 22Na+ as we previously described (Cai, Wu et al. 2008). Cells were

pretreated with 50 µM amiloride to inhibit amiloride-sensitive NHE1 activity.

4.3.5 Measurement of c-Src phosphorylation

127 Whole cell lysates were prepared with Nonidet P-40 buffer containing 1%

Nonidet P-40, 0.25% sodium deoxycholate, 50 mM NaCl, 50 mM HEPES, 10%

Glycerol (pH 7.4), 1 mM sodium vanadate, 0.5 mM sodium fluoride, 1 mM

Phenylmethanesulfonyl fluoride, and protease inhibitor cocktail for general use

(Sigma). After clarification, 300μg of total protein was immunoprecipitated with

antibody against c-Src and protein G-agrose beads (Millipore), and then eluted with

2x Laemmli buffer. After immunoblotting for phospho-c-Src (p-Src), the same

membrane was stripped and immunoblotted for total c-Src (t-Src). The activation

(phosphorylation) of c-Src was expressed as ratio of p-Src/ t-Src with both

measurements normalized to 1.0 for the control samples (Liu, Yan et al. 2011).

4.3.6 Dihydroethidium staining

Intracellular superoxide levels are determined qualitatively by

- dihydroethidium (DHE) fluorescence, a specific probe for intracellular O2 production (Beswick, Dorrance et al. 2001; Elmarakby, Loomis et al. 2005;

Robinson, Janes et al. 2008; Cossarizza, Ferraresi et al. 2009). DHE is a lipophilic cell-permeable dye that is rapidly oxidized to fluorescent product ethidium in the presence of free radical superoxide. Produced ethidium tends to intercalate into nuclear DNA and fluoresces strongly at around 600 nm when excited at 500–530 nm, giving an indication of oxidant stress. Cells were loaded with 5 μM dihydroethidium (DHE) for 20 min. After washing 2 times in phosphate- buffered saline (PBS), cells were scanned for DHE red fluorescence using an Olympus

128 FSX100 box type fluorescence imaging device (Olympus America, Center Valley,

PA). The DHE excitation wavelength is 488nm and emission wavelength is 585nm.

4.3.7 Assessment of formation of DMPO-nitrone adducts (Spin-trap

immunoassay or Immuno-spin trapping)

The nitrone spin trap 5,5-dimethyl-1-5 pyrroline N-oxide (DMPO) is widely used

to measure protein-centered radicals (Ramirez 2005). In theory, protein-centered radicals might be trapped with the nitrone spin trap DMPO forming DMPO-radical adducts, which can be detected by using an antibody. LLC-PK1 cells were pretreated with DMPO

(50mM) for 1 h and then with ouabain or glucose oxidase for 1h at concentration indicated. Whole cell lysates were prepared with Nonidet P-40 buffer as aforementioned, separated by SDS-PAGE, and immunoblotted with anti-DMPO antibody.

4.3.8 Assessment of protein carbonylation

Whole cell lysates were prepared as aforementioned. Equal amount of proteins from each sample was denatured with 6% SDS (final concentration), derivatized with 1x DNPH (diluted freshly with distilled water from 10x DNPH stock solution, 100mM in 100% Trifluoroacetic acid), and neutralized with neutralization Buffer (30% of Glycerol in 2M Tris). This was followed by either

Western blot for protein carbonylation in whole cell lysates or immunoprecipitation

(IP-DNP) studies. For IP-DNP, neutralized DNP-derivatives were pull-down with anti-DNP antibody (Sigma) and protein G-agrose beads, and then eluted with 2x

129 Laemmli buffer. Elutes were immunoblotted with antibodies against Na/K-ATPase

α1 subunit, NHE3 and c-Src.

4.3.9 Protein Identification by LC-MS/MS

Protein identification was performed in Mass Spectrometry-based Proteomics

Facility (Department of Pathology, University of Michigan), based on previously

described protocols (Todi, Scaglione et al.). LLC-PK1 cells were treated with

ouabain (100nM) or GO (3mU/ml) for 1 hr. The Na/K-ATPase α1 subunit was

immunoprecipitated with anti-α1 antibody and separated with SDS-PAGE.

Coomassie Brilliant Blue staining was performed using mass spectrometry-

compatible NOVEX Coomassie Blue colloidal staining (Invitrogen) as instructed by

the manufacturer. The α1 bands were exercised and processed for in-gel trypsin

digestion with sequencing grade modified trypsin (Promega). Resulting peptides

were resolved on a nano-capillary reverse phase column (Picofrit column, New

Objective) and directly introduced into a linear ion-trap mass spectrometer (LTQ

Orbitrap XL, Thermo Fisher). Data-dependent MS/MS spectra on the five most

intense ions from each full MS scan were collected (relative Collision Energy

~35%). Proteins were identified by searching the data against database appended

with decoy (reverse) sequences using the X!Tandem/Trans-Proteomic Pipeline

(TPP) software suite. All peptides and proteins with a PeptideProphet and

ProteinProphet probability score of >0.85 (false discovery rate <3%) were

considered positive identifications of direct protein carbonylation manually verified.

130 4.3.8 Western blotting

Equal amounts of total protein were resolved by 10% sodium dodecylsulfate-

polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to PVDF

membrane. For protein carbonylation assay, the membrane was stained using

Ponceau S solution (Sigma P7171-1L) to verify uniform loading before

immunoblotting with indicated antibodies. Signal detection was performed with an

enhanced chemiluminescence (ECL) super signal kit (Pierce, Rockford, IL).

Multiple exposures were analyzed to assure that the signals were within the linear

range of the film. The signal density was determined using Molecular Analyst

software (Bio-Rad, Hercules, CA).

4.3.9 Statistical analysis

Data were tested for normality (all data passed) and then subjected to parametric analysis. When more than two groups were compared, one-way analysis

of variance (ANOVA) was performed prior to comparison of individual groups, and

the post-hoc t-tests were adjusted for multiple comparisons using Bonferroni’s

correction. Statistical significance was reported at the P < 0.05 and P < 0.01 levels.

SPSS software was used for all analysis. Values are given as mean±SEM.

4.4 Results

4.4.1 The effect of ROS on ouabain-initiated Na/K-ATPase/c-Src signaling

131 In LLC-PK1 cells, we have demonstrated that the Na/K-ATPase α1 subunit directly interacts with c-Src kinase via two pairs of domain interactions to form a functional receptor complex, and ouabain stimulated phosphorylation of c-Src bound to the α1 subunit and association between the α1 subunit and c-Src (Tian, Cai et al.

2006; Li and Xie 2009). We determined the role of ROS on ouabain-stimulated c-Src phosphorylation in LLC-PK1 cells. As shown in Fig. 4-1, ouabain (100nM, 15min) stimulated c-Src, ERK1/2 activation and interaction between the α1 subunit and c-

Src, which was abolished by pretreatment with NAC (N-Acetyl-L-cysteine, 10mM,

30min) (Fig. 4-1a-d). NAC alone has no effect on c-Src activation. Cobalt protoporphyrin (CoPP), heme oxygenase (HO-1) inducer (antioxidant enzyme), attenuated ouabain stimulated-c-Src activation (Fig. 4-1e-f). Binding of ouabain to the Na/K-ATPase not only recruits c-Src to the signaling complex (Haas, Wang et al.

2002), but it also activates c-Src that binds to the Na/K-ATPase α1 subunit (Tian,

Cai et al. 2006). Since NAC is capable of eliminating basal ROS before ouabain stimulation, it appears that a basal level of ROS is required, as a co-effector, in ouabain-induced Na/K-ATPase/c-Src signaling in which, a basal level of ROS might affect ouabain binding to the α1 subunit by induction of Na/K-ATPase conformation change, favoring ouabain binding and c-Src activation.

4.4.2 The effect of ROS on ouabain-induced redistribution of the α1 subunit and

NHE3, and inhibition of transcellular 22Na+ flux

Ouabain and a high salt diet redistribute the Na/K-ATPase and NHE3 to promote renal sodium excretion by signaling through the Na/K-ATPase (Cai, Wu et

132 al. 2008; Liu, Yan et al. 2011). Consistent with the observation that NAC prevents

ouabain-induced c-Src, ERK1/2 activation (Fig. 4-1a-d), pretreatment with NAC

(10mM, 30min) also prevented ouabain (100nM, 1h) stimulated accumulation of

Na/K-ATPase α1 and NHE3 in early endosome (EE) fractions (Fig. 4-2a-c). The

Na/K-ATPase in rat cerebellar granule cells is redox-sensitive with a "optimal redox

potential range" (Petrushanko, Bogdanov et al. 2006). Interestingly, ouabain-induced

c-Src activation was significantly attenuated by pretreatment (30min) with high

doses but not low dose of NAC in LLC-PK1 cells (Fig. 4-2e). Functionally, ouabain

(100nM, 1h)-induced inhibition of active transepithelial 22Na+ flux was blunted by

pretreatment of 10mM NAC (30min), but not 1mM NAC (Fig. 4-2f). Taken together, the above data further supported our hypothesis that a certain level of ROS is

required in ouabain-induced Na/K-ATPase/c-Src signaling and redistribution of the

Na/K-ATPase and NHE3, which lead to inhibition of sodium reabsorption in renal

proximal tubules.

4.4.3 Ouabain stimulated ROS production and protein oxidation

- DHE (dihydroethidine) is extensively used as a probe for O2 , being oxidized

to a fluorescent product to evaluate ROS production. As shown in Fig. 4-3a, ouabain

(100nM, 1h) induced ROS production was assessed by DHE staining. In evaluating

protein oxidation studies, we use GO (glucose oxidase), which causes a steady

increase in intracellular ROS in cultured cardiac myocytes for at least 12 h (Liu,

Tian et al. 2000; Liu, Li et al. 2006), as positive control. Both ouabain (50 and

100nM, 1h) and GO (1 and 3 mU/ml, 1h) stimulated the formation of DMPO (5,5-

133 Dimethyl-1-Pyrroline-N-Oxide)-nitrone adducts assessed by a spin trap reagent

DMPO-based Western blot assay (Fig. 4-3b). Cells with DMPO treatment alone served as system control. Ouabain (100nM, 1h) and GO (3 mU/ml, 1h) also stimulated protein carbonylation in whole cell lysates (Fig. 4-3c). When whole cell lysate proteins were derivatized with DNPH (2,4-Dinitrophenylhydrazine) and immunoprecipitated with anti-DNP antibody, it appeared that both ouabain (100nM,

1h) and GO (3mU/ml, 1h) caused protein carbonylation of Na/K-ATPase α1 subunit,

NHE3 and c-Src (Fig. 4-3d). When whole cell lysate proteins were immunoprecipitated with anti-α1 subunit antibody and then derivatized with DNPH, similar carbonylation of the α1 subunit stimulated by ouabain and GO was observed

(data not shown). In Fig. 4-3b-c, control immunoblots with DMPO or DNPH alone suggest the presence of basal ROS and protein oxidation.

These results led us to speculate that oxidative modification of the α1 subunit might promote its internalization. We compared the protein carbonylation profiles in whole cell lysate and EE fraction. Equal amount of total proteins (20µg) was used to determine the protein carbonylation profiles. As shown in Fig. 4-3e, the profile of whole cell lysate is significantly different from that of EE fractions. The most profound difference is the band closely located between 100-115 kDa that is presented as the α1 subunit, as indicated in Fig. 4-3d.

134 4.4.4 ROS stress stimulated Na/K-ATPase signaling and redistribution of Na/K-

ATPase and NHE3, and inhibited transepithelial 22Na+ flux

It has been shown that oxidative stress induces conformation change and

inhibits Na/K-ATPase activity (Huang, Wang et al. 1992; Figtree, Liu et al. 2009).

Like ouabain (100nM, 15min), GO (3mU/ml, 15min) alone stimulated

phosphorylation of c-Src and ERK1/2 (Fig. 4-4a-b). Moreover, co-treatment with

ouabain (100nM) and GO (3mU/ml) for 15min had a slight but significant

synergistic effects on c-Src activation compared to ouabain or GO alone (Fig. 4-4a),

further indicating that ROS is a co-effector of ouabain-induced Na/K-ATPase/c-Src

signaling. The data also suggest that ouabain-induced ROS generation, functioning

as a downstream effector of Na/K-ATPase/c-Src signaling, might further stimulate

Na/K-ATPase/c-Src cascade by a positive feedback mechanism. Increases in ROS

stress by GO (1 and 3mU/ml) alone activated c-Src (Fig. 4-4c, 15min treatment) and

stimulated Na/K-ATPase endocytosis (Fig. 4-4c-e, 1h treatments). In transepithelial

22Na+ flux assay, GO (3mU/ml, 60 min) alone, like ouabain (100nM, 60min),

inhibited the 22Na+ flux. However, unlike ouabain-induced inhibition only occurred when ouabain was applied in the basolateral aspect (Cai, Wu et al. 2008), GO inhibited the transepithelial 22Na+ flux when it was applied on both basolateral and

apical aspects (Fig. 4-4f). Taken together, the data indicate that GO alone can

activate c-Src (Fig. 4-4a-c), stimulate protein oxidative modification (Fig. 4-3b-d)

and the redistribution of the Na/K-ATPase (Fig. 4-4c, e) in the same manner as

ouabain. This is consistent with out early observations that ROS induced by GO

135 crosstalk with the Na/K-ATPase and induce the Na/K-ATPase endocytosis (Liu, Li

et al. 2006).

4.4.5 The role of c-Src in ouabain and glucose oxidase mediated protein

carbonylation.

Because c-Src activation is the initiating step in Na/K-ATPase signaling

cascade, to obtain additional evidence showing the interplay between ROS and

Na/K-ATPase signaling, we investigated whether ROS generation is influenced by

c-Src. LLC-PK1 cells were pretreated with c-Src specific inhibitor PP2, and then

exposed to 100nM ouabain or 3mU/ml GO to assess c-Src activation, ERK1/2

activation, as well as protein carbonylation. As depicted in Fig. 4-5a-c, PP2

abrogated c-Src and ERK1/2 phosphorylation induced by GO. Interestingly, PP2

abolished ouabain-induced protein carbonylation and attenuated GO-mediated

protein oxidation. These data support our previous propositions that Na/K-ATPase

signaling is implicated in ROS generation and c-Src plays a key role in ouabain-

induced protein oxidation. Given PP2 is non-specific kinase inhibitor, both SYF and

SYF+c-Src cells were employed to further demonstrate the hypothesis that c-Src is

necessary in ROS generation caused by ouabain. The SYF+c-Src cells are derived

from mouse embryos harboring functional null mutations in both alleles of the c-Src

family kinases Src, Yes, and Fyn. The SYF+c-Src cells are the stable transfectants of

SYF cells that express c-Src (Haas, Wang et al. 2002). As shown in Fig. 4-5d-e, in

SYF cells, protein carbonylation was not significantly altered by ouabain compared

with controls. However, in SYF+c-Src cells, ouabain significantly increased protein

136 carbonylation. Glucose oxidase caused protein oxidation in both SYF and SYF+c-

Src cells. The data suggest c-Src is required in ROS generation mediated by ouabain, while glucose oxidase-induced protein oxidation is partially dependent on c-Src, and

Na/K-ATPase might not be the only pathway for GO-mediated protein carbonylation.

4.4.6 Disruption of Na/K-ATPase/c-Src signaling attenuated ouabain-induced

ROS production

To test the role of Na/K-ATPase/c-Src complex in ouabain-induced ROS generation, we used two stable cell lines generated from LLC-PK1 by siRNA method, with disrupted Na/K-ATPase/c-Src signaling and signaling complex. PY-17 cells only express about 8-10% of α1 compared to the parent LLC-PK1, and C2-9 cells are caveolin-1 knock-out LLC-PK1 cells showing disrupted formation of caveolar Na/K-ATPase/c-Src complex (Liu, Liang et al. 2005; Liang, Cai et al. 2006;

Liang, Tian et al. 2007). The basal level of c-Src phosphorylation is higher in PY-17 cells than that in LLC-PK1 cells (Liang, Cai et al. 2006; Liang, Tian et al. 2007).

Both PY-17 and C2-9 do not respond to ouabain stimulation in terms of c-Src activation. Furthermore, depletion of caveolin-1 or disruption of lipid rafts abolishes ouabain-induced endocytosis of the Na/K-ATPase (Liu, Liang et al. 2005; Cai, Wu et al. 2008). The ouabain-induced c-Src activation (ouabain=100nM, 15min) (Fig. 4-

6a) and ROS production (ouabain=100nM, 1h) (Fig. 4-6b-c) were significantly attenuated by disruption of the Na/K-ATPase/c-Src signaling complex. Since ouabain-induced redistribution of the Na/K-ATPase and NHE3 requires caveolin-1 and lipid rafts (Liu, Liang et al. 2005; Cai, Wu et al. 2008), the data suggest that

137 ouabain-induced ROS generation requires Na/K-ATPase/c-Src signaling, and ouabain-induced protein oxidation is a downstream effector of Na/K-ATPase/c-Src signaling (as shown in Fig. 4-5c-e and Fig. 4-6b-c) to promote the redistribution of certain proteins in EE fractions (Fig. 4-3e).

4.4.7 Identification of carbonylation sites in the Na/K-ATPase α1 subunit

To determine the carbonylation site(s), LC-MS/MS was conducted with native trypsinized α1 subunit isolated from LLC-PK1 cells by immunoprecipitation, with or without treatment with ouabain (100nM, 1 hr) or GO (3mU/ml, 1hr). Direct carbonylation modification of two amino acid residues Pro234 (mass 113.0477 with mass diff 15.9949) and Thr237 (mass 99.0320 with mass diff -2.0156) in peptide

226SPDFTNENPLETR238 (numbered by UniProtKB/Swiss-Prot No P05024

(AT1A1_PIG)) were present in control ouabain- and GO- treated cells. Both Pro224 and Thr226 in peptide 211VDNSSLTGESEPQTR225 were shown stimulated direct carbonylation only in response to ouabain and GO treatments (Fig. 4-7). Both peptides have 2 tolerable (tryptic) termini (NTT=2). Comparing to peptide

226SPDFTNENPLETR238, peptide 211VDNSSLTGESEPQTR225 is highly conserved and present in Na/K-ATPase α1/α2/α3/α4 isoforms. Moreover, these two peptides are integrity linked (211VDNSSLTGESEPQTRSPDFTNENPLETR238) and located in the actuator (A) domain (cytosolic loop between transmembrane M2 and M3 domains) of the α1 subunit, and Pro222/Thr224 is located on the surface facing the nucleotide binding (N) domain. Upon ouabain binding, the Na/K-ATPase undergoes conformation changes, in which the A domain is rotated to the N domain and

138 transmembrane M1/M2 domain is shifted toward M3/M4 domain (Yatime, Laursen

et al.). These conformation changes form a high affinity binding pocket for ouabain

as well as might stabilize caveolin-1 binding motif and affect binding of signaling

molecules such as c-Src, PI3K and IP3R (Yatime, Laursen et al.), which are critical

in the Na/K-ATPase signaling. Except the two carbonylation sites, the peptide

211VDNSSLTGESEPQTR225 also includes 217TGES220 motif and Glu221 residue that

affect conformation change and function. The 217TGES220 motif not only is the

anchor motif of A domain rotated toward N domain (~10˚), but it also protects spontaneous hydrolysis of phosphorylated Asp374 residue in E2P:Ouabain state

(Yatime, Laursen et al.). Moreover, Glu221 and Arg549 form the only link between the

A and N domains (Toyoshima, Kanai et al.). However, it is not clear the effect of

Pro224/Thr226 carbonylation on the link between Glu221 and Arg549.

4.5 Discussion

We have shown that activation of the Na/K-ATPase/c-Src signaling by a high

salt diet or ouabain inhibits RPT sodium reabsorption to counterbalance salt-loading

mediated volume expansion and related BP increase. We reported here that in LLC-

PK1 cells, ROS and ouabain-Na/K-ATPase/c-Src signaling are inextricably linked

forming a positive feedback mechanism. Basal ROS is required to initiate and

propagate the Na/K-ATPase signaling. Neutralization of ROS with NAC prevents

ouabain-induced activation of Na/K-ATPase/c-Src signaling, redistribution of Na/K-

ATPase and NHE3, and inhibition of transcellular 22Na+ flux. Furthermore, ouabain-

139 stimulated ROS generation might link the Na/K-ATPase signaling to NHE3 regulation.

It was first proposed that an increase in endogenous CTS stimulates renal sodium excretion by inhibition of renal tubular Na/K-ATPase to correct volume expansion and its related BP increase (Haddy, Pamnani et al. 1979; de Wardener and Clarkson 1985), but the finding that ouabain mediated vascular tone shifts the focus away from renal function (Blaustein, Zhang et al. 2009). Gene replacement studies have unequivocally demonstrated an important role of endogenous CTS in regulation of renal sodium excretion and BP (Dostanic-Larson, Van Huysse et al.

2005; Loreaux, Kaul et al. 2008). Recently, evidences including ours support this concept under pathological conditions such as high salt intake, chronic renal failure and chronic heart failure in various animal models and human beings. We have demonstrated that the Na/K-ATPase α1 subunit directly interacts with c-Src kinase via two pairs of domain-domain interactions to form a functional receptor complex

(Tian, Cai et al. 2006; Li and Xie 2009), as well as that CTS activate this receptor

Na/K-ATPase/c-Src complex, resulting in the stimulation of multiple protein kinase cascades including c-Src, ERK1/2, PI3K and PKC, and increased ROS production in both in vitro and in vivo models (reviewed in (Schoner and Scheiner-Bobis 2008;

Bagrov, Shapiro et al. 2009; Li and Xie 2009; Liu and Xie 2010)). Ouabain- stimulated ROS generation functions as a second messenger in ouabain-activated

Na/K-ATPase signaling (Liu, Tian et al. 2000; Liu, Li et al. 2006). Infusion of CTS, like angiotensin II, causes ROS generation and protein oxidation in experimental animals (Kennedy, Vetteth et al. 2006; Kennedy, Vetteth et al. 2006). Oxidative

140 modification of the Na/K-ATPase inhibits its activity, promotes its susceptibility to

degradation, and stabilizes the enzyme in an E2-prone conformation (Xie, Wang et al. 1990; Huang, Wang et al. 1992; THÉVENOD and FRIEDMANN 1999; Figtree,

Liu et al. 2009). An increase in ROS stimulates interaction between the Na/K-

ATPase and signaling proteins which leads to trafficking regulation of the Na/K-

ATPase (Liu, Li et al. 2006; Dada, Novoa et al. 2007). Our present study indicated

that ouabain binding stimulated carbonylation modification of the Na/K-ATPase α1

subunit and NHE3 might trigger the trafficking regulation.

Increases in oxidative stress are both a cause and consequence of

hypertension (Touyz 2004; Wilcox 2005; Vaziri and Rodriguez-Iturbe 2006; Welch

2006). High salt diet, which is well-documented for its stimulation of systematic

oxidative stress, regulates the activity and distribution of the Na/K-ATPase and

NHE3 that contribute to the development and maintenance of hypertension in

different animal models (Moe, Tejedor et al. 1991; Silva and Soares-da-Silva 2007;

Panico, Luo et al. 2009; McDonough 2010; Banday and Lokhandwala 2011; Liu,

Yan et al. 2011). In the kidney, increased oxidative stress influences a number of

physiologic processes including renal sodium handling in the proximal tubule

(Zhang, Imam et al. 2002; Han, Lee et al. 2005; Panico, Luo et al. 2009; Schreck

and O'Connor 2011), and contributes to salt-sensitivity and salt-sensitive

hypertension (Kopkan and Majid 2005; Taylor, Glocka et al. 2006). Anti-oxidant

agents such as Tempol and enzymes such as heme oxygenase-1 exhibit a beneficial

and protective effect on BP in various animal models of hypertension (Abraham

and Kappas 2005). However, the beneficial effect of antioxidants is controversy and

141 not seen in most clinic trials with administration of antioxidant supplementation

(reviewed in (Touyz 2004; Munzel, Gori et al. 2010)). It is suggested that synthetic

antioxidant supplementation may be ineffective or even dangerous due to the

possible ‘over-antioxidant-buffering’ effect of excessive antioxidant

supplementation (Huang, Caballero et al. 2006), while natural antioxidants from

high dietary intakes of fruits, vegetables, nuts and grains might significantly lower

systolic BP (Kizhakekuttu and Widlansky ; John, Ziebland et al. 2002). It appears

that maintaining a proper range of ROS, systematically and locally, is more

important.

Carbonylation modifications, via metal-catalyzed activation of hydrogen

peroxide, are very stable products and chemically irreversible, and carbonylated

proteins are prone to be degradated (Stadtman and Levine 2000; Dalle-Donne,

Aldini et al. 2006). However, a recent study has demonstrated the possible role of

carbonylation/decarbonylation process in signal transduction in which the thiol

groups were responsible for decarbonylation via enzymatic processes in the

biologic system (Wong, Marcocci et al. 2010). Our observations suggest that

carbonylated proteins were gradually recovered after removal of un-bound ouabain

(data not shown). Although the task of identifying the causal factors behind time-

dependent decreased carbonylation has been proven difficult, several principle

possibilities can be envisioned, including (1) a de novo protein synthesis to dilute

carbonylated protein levels, (2) degradation for removal of oxidized proteins, (3) decarbonylation. To our knowledge, this is the first time to demonstrate that CTS and/or ROS cause direct carbonylation modification, in the A domain that has the

142 potential to significantly affect Na/K-ATPase α1 conformation change and known binding domains for partners of the Na/K-ATPase signaling. Nonetheless, more studies are needed to explore the underlying mechanism.

Our present data indicate a novel mechanism regulating RPT sodium handling via interaction of ROS with the Na/K-ATPase/c-Src signaling pathway, which may influence salt retention related volume expansion that is responsible for certain types of hypertension. However, the mechanisms remain largely to be elucidated since the available data is limited (Garvin and Ortiz 2003; Schreck and

O'Connor 2011). Some pertinent questions remain to be resolved, such as how ROS interacts with and influences the Na/K-ATPase/c-Src signaling cascade as well as whether ouabain-induced ROS acutely boosts the Na/K-ATPase signaling by a positive feedback mechanism and chronically desensitizes the signaling cascade by stimulating Na/K-ATPase/c-Src endocytosis.

Acknowledgments

The authors would like to thank Dr. Venkatesha Basrur (Department of

Pathology, University of Michigan) for performing MASS analysis and Ms. Carol

Woods for her excellent help. Portions of this work were supported by grants from

National Institutes of Health (HL-109015 to Z.X. and J.I.S., and GM-78565 to Z.X.).

143 4.6 References

Abraham, N. G. and A. Kappas (2005). "Heme oxygenase and the cardiovascular-renal

system." Free Radic Biol Med 39(1): 1-25.

Bagrov, A. Y., J. I. Shapiro, et al. (2009). "Endogenous cardiotonic steroids: physiology,

pharmacology, and novel therapeutic targets." Pharmacol Rev 61(1): 9-38.

Banday, A. A. and M. F. Lokhandwala (2011). "Angiotensin II-mediated biphasic

regulation of proximal tubular Na+/H+ exchanger 3 is impaired during oxidative

stress." Am J Physiol Renal Physiol 301(2): F364-70.

Beswick, R. A., A. M. Dorrance, et al. (2001). "NADH/NADPH oxidase and enhanced

superoxide production in the mineralocorticoid hypertensive rat." Hypertension

38(5): 1107-11.

Blaustein, M. P., J. Zhang, et al. (2009). "The pump, the exchanger, and endogenous

ouabain: signaling mechanisms that link salt retention to hypertension."

Hypertension 53(2): 291-8.

Cai, H., L. Wu, et al. (2008). "Regulation of apical NHE3 trafficking by ouabain-induced

activation of the basolateral Na+-K+-ATPase receptor complex." Am J Physiol

Cell Physiol 294(2): C555-63.

Calhoun, D. A., D. Jones, et al. (2008). "Resistant Hypertension: Diagnosis, Evaluation,

and Treatment: A Scientific Statement From the American Heart Association

Professional Education Committee of the Council for High Blood Pressure

Research." Hypertension 51(6): 1403-1419.

144 Cossarizza, A., R. Ferraresi, et al. (2009). "Simultaneous analysis of reactive oxygen

species and reduced glutathione content in living cells by polychromatic flow

cytometry." Nat Protoc 4(12): 1790-7.

Dada, L. A., E. Novoa, et al. (2007). "Role of the small GTPase RhoA in the hypoxia-

induced decrease of plasma membrane Na,K-ATPase in A549 cells." J Cell Sci

120(Pt 13): 2214-22.

Dalle-Donne, I., G. Aldini, et al. (2006). "Protein carbonylation, cellular dysfunction, and

disease progression." J Cell Mol Med 10(2): 389-406.

Dalle-Donne, I., R. Rossi, et al. (2003). "Protein carbonyl groups as biomarkers of

oxidative stress." Clin Chim Acta 329(1-2): 23-38.

de Wardener, H. E. and E. M. Clarkson (1985). "Concept of natriuretic hormone."

Physiological Reviews 65(3): 658-759.

Dostanic-Larson, I., J. W. Van Huysse, et al. (2005). "The highly conserved cardiac

glycoside binding site of Na,K-ATPase plays a role in blood pressure regulation."

Proc Natl Acad Sci U S A 102(44): 15845-50.

Elmarakby, A. A., E. D. Loomis, et al. (2005). "NADPH oxidase inhibition attenuates

oxidative stress but not hypertension produced by chronic ET-1." Hypertension

45(2): 283-7.

Fedorova, O. V., J. I. Shapiro, et al. (2010). "Endogenous cardiotonic steroids and salt-

sensitive hypertension." Biochim Biophys Acta 1802(12): 1230-6.

Figtree, G. A., C.-C. Liu, et al. (2009). "Reversible Oxidative Modification: A Key

Mechanism of Na+-K+ Pump Regulation." Circ Res 105(2): 185-193.

145 Garvin, J. L. and P. A. Ortiz (2003). "The role of reactive oxygen species in the

regulation of tubular function." Acta Physiol Scand 179(3): 225-32.

Gorvel, J. P., P. Chavrier, et al. (1991). "rab5 controls early endosome fusion in vitro."

Cell 64(5): 915-25.

Haas, M., H. Wang, et al. (2002). "Src-mediated inter-receptor cross-talk between the

Na+/K+-ATPase and the epidermal growth factor receptor relays the signal from

ouabain to mitogen-activated protein kinases." J Biol Chem 277(21): 18694-702.

Haddy, F. J., M. B. Pamnani, et al. (1979). "Humoral factors and the sodium-potassium

pump in volume expanded hypertension." Life Sci 24(23): 2105-17.

Han, H. J., Y. J. Lee, et al. (2005). "High glucose-induced oxidative stress inhibits

Na+/glucose cotransporter activity in renal proximal tubule cells." American

Journal of Physiology - Renal Physiology 288(5): F988-F996.

Huang, H.-Y., B. Caballero, et al. (2006). "The Efficacy and Safety of Multivitamin and

Mineral Supplement Use To Prevent Cancer and Chronic Disease in Adults: A

Systematic Review for a National Institutes of Health State-of-the-Science

Conference." Annals of Internal Medicine 145(5): 372-385.

Huang, W. H., Y. Wang, et al. (1992). "(Na+ + K+)-ATPase: inactivation and

degradation induced by oxygen radicals." Int J Biochem 24(4): 621-6.

John, J. H., S. Ziebland, et al. (2002). "Effects of fruit and vegetable consumption on

plasma antioxidant concentrations and blood pressure: a randomised controlled

trial." Lancet 359(9322): 1969-74.

146 Kennedy, D. J., S. Vetteth, et al. (2006). "Central role for the cardiotonic steroid

marinobufagenin in the pathogenesis of experimental uremic cardiomyopathy."

Hypertension 47(3): 488-95.

Kennedy, D. J., S. Vetteth, et al. (2006). "Ouabain decreases sarco(endo)plasmic

reticulum calcium ATPase activity in rat hearts by a process involving protein

oxidation." Am J Physiol Heart Circ Physiol 291(6): H3003-11.

Kizhakekuttu, T. J. and M. E. Widlansky "Natural antioxidants and hypertension:

promise and challenges." Cardiovasc Ther 28(4): e20-32.

Kopkan, L. and D. S. A. Majid (2005). "Superoxide Contributes to Development of Salt

Sensitivity and Hypertension Induced by Nitric Oxide Deficiency." Hypertension

46(4): 1026-1031.

Li, Z. and Z. Xie (2009). "The Na/K-ATPase/Src complex and cardiotonic steroid-

activated protein kinase cascades." Pflugers Arch 457(3): 635-44.

Liang, M., T. Cai, et al. (2006). "Functional characterization of Src-interacting Na/K-

ATPase using RNA interference assay." J Biol Chem 281(28): 19709-19.

Liang, M., J. Tian, et al. (2007). "Identification of a pool of non-pumping Na/K-ATPase."

J Biol Chem 282(14): 10585-93.

Liu, J., R. Kesiry, et al. (2004). "Ouabain induces endocytosis of plasmalemmal Na/K-

ATPase in LLC-PK1 cells by a clathrin-dependent mechanism." Kidney Int 66(1):

227-41.

Liu, J., M. Liang, et al. (2005). "Ouabain-induced endocytosis of the plasmalemmal

Na/K-ATPase in LLC-PK1 cells requires caveolin-1." Kidney Int 67(5): 1844-54.

147 Liu, J., J. Tian, et al. (2000). "Ouabain interaction with cardiac Na+/K+-ATPase initiates

signal cascades independent of changes in intracellular Na+ and Ca2+

concentrations." J Biol Chem 275(36): 27838-44.

Liu, J. and Z. J. Xie (2010). "The sodium pump and cardiotonic steroids-induced signal

transduction protein kinases and calcium-signaling microdomain in regulation of

transporter trafficking." Biochim Biophys Acta 1802(12): 1237-45.

Liu, J., Y. Yan, et al. (2011). "Impairment of Na/K-ATPase Signaling in Renal Proximal

Tubule Contributes to Dahl Salt-sensitive Hypertension." J Biol Chem 286(26):

22806-13.

Liu, L., J. Li, et al. (2006). "Involvement of Na+/K+-ATPase in hydrogen peroxide-

induced hypertrophy in cardiac myocytes." Free Radic Biol Med 41(10): 1548-56.

Loreaux, E. L., B. Kaul, et al. (2008). "Ouabain-Sensitive alpha1 Na,K-ATPase enhances

natriuretic response to saline load." J Am Soc Nephrol 19(10): 1947-54.

McDonough, A. A. (2010). "Mechanisms of proximal tubule sodium transport regulation

that link extracellular fluid volume and blood pressure." American Journal of

Physiology - Regulatory, Integrative and Comparative Physiology 298(4): R851-

R861.

Moe, O. W., A. Tejedor, et al. (1991). "Dietary NaCl modulates Na(+)-H+ antiporter

activity in renal cortical apical membrane vesicles." American Journal of

Physiology - Renal Physiology 260(1): F130-F137.

Mokry, M. and E. Cuppen (2008). "The Atp1a1 gene from inbred Dahl salt sensitive rats

does not contain the A1079T missense transversion." Hypertension 51(4): 922-7.

148 Munzel, T., T. Gori, et al. (2010). "Is oxidative stress a therapeutic target in

cardiovascular disease?" Eur Heart J 31(22): 2741-8.

Nishi, A., A. M. Bertorello, et al. (1993). "Renal Na+,K(+)-ATPase in Dahl salt-sensitive

rats: K+ dependence, effect of cell environment and protein kinases." Acta

Physiol Scand 149(3): 377-84.

Oweis, S., L. Wu, et al. (2006). "Cardiac glycoside downregulates NHE3 activity and

expression in LLC-PK1 cells." Am J Physiol Renal Physiol 290(5): F997-1008.

Panico, C., Z. Luo, et al. (2009). "Renal proximal tubular reabsorption is reduced in adult

spontaneously hypertensive rats: roles of superoxide and Na+/H+ exchanger 3."

Hypertension 54(6): 1291-7.

Periyasamy, S. M., J. Liu, et al. (2005). "Salt loading induces redistribution of the

plasmalemmal Na/K-ATPase in proximal tubule cells." Kidney Int 67(5): 1868-77.

Petrushanko, I., N. Bogdanov, et al. (2006). "Na-K-ATPase in rat cerebellar granule cells

is redox sensitive." American Journal of Physiology - Regulatory, Integrative and

Comparative Physiology 290(4): R916-R925.

Ramirez, D. a. M., RP (2005). "Immuno-spin trapping: detection of protein-centered

radicals." in Current Protocols in Toxicology, L. Costa, Maines, MD, Reed, DJ,

Sassa, S. and Sipes, IG, Editor.: p. 17.7.1–17.7.16.

Robinson, K. M., M. S. Janes, et al. (2008). "The selective detection of mitochondrial

superoxide by live cell imaging." Nat Protoc 3(6): 941-7.

Schoner, W. and G. Scheiner-Bobis (2008). "Role of endogenous cardiotonic steroids in

sodium homeostasis." Nephrol Dial Transplant 23(9): 2723-9.

149 Schreck, C. and P. M. O'Connor (2011). "NAD(P)H oxidase and renal epithelial ion

transport." American Journal of Physiology - Regulatory, Integrative and

Comparative Physiology 300(5): R1023-R1029.

Silva, E. and P. Soares-da-Silva (2007). "Reactive oxygen species and the regulation of

renal Na+-K+-ATPase in opossum kidney cells." American Journal of Physiology

- Regulatory, Integrative and Comparative Physiology 293(4): R1764-R1770.

Stadtman, E. R. and R. L. Levine (2000). "Protein oxidation." Ann N Y Acad Sci 899:

191-208.

Stadtman, E. R. and R. L. Levine (2003). "Free radical-mediated oxidation of free amino

acids and amino acid residues in proteins." Amino Acids 25(3-4): 207-18.

Taylor, N. E., P. Glocka, et al. (2006). "NADPH oxidase in the renal medulla causes

oxidative stress and contributes to salt-sensitive hypertension in Dahl S rats."

Hypertension 47(4): 692-8.

THÉVENOD, F. and J. M. FRIEDMANN (1999). "Cadmium-mediated oxidative stress

in kidney proximal tubule cells induces degradation of Na+/K+-ATPase through

proteasomal and endo-/lysosomal proteolytic pathways." The FASEB Journal

13(13): 1751-1761.

Tian, J., T. Cai, et al. (2006). "Binding of Src to Na+/K+-ATPase forms a functional

signaling complex." Mol Biol Cell 17(1): 317-26.

Todi, S. V., K. M. Scaglione, et al. "Activity and cellular functions of the

deubiquitinating enzyme and polyglutamine disease protein ataxin-3 are regulated

by ubiquitination at lysine 117." J Biol Chem 285(50): 39303-13.

150 Touyz, R. M. (2004). "Reactive oxygen species, vascular oxidative stress, and redox

signaling in hypertension: what is the clinical significance?" Hypertension 44(3):

248-52.

Toyoshima, C., R. Kanai, et al. "First crystal structures of Na+,K+-ATPase: new light on

the oldest ion pump." Structure 19(12): 1732-8.

Vaziri, N. D. and B. Rodriguez-Iturbe (2006). "Mechanisms of disease: oxidative stress

and inflammation in the pathogenesis of hypertension." Nat Clin Pract Nephrol

2(10): 582-93.

Welch, W. J. (2006). "Intrarenal oxygen and hypertension." Clin Exp Pharmacol Physiol

33(10): 1002-5.

Wilcox, C. S. (2005). "Oxidative stress and nitric oxide deficiency in the kidney: a

critical link to hypertension?" Am J Physiol Regul Integr Comp Physiol 289(4):

R913-35.

Wong, C. M., L. Marcocci, et al. (2010). "Cell signaling by protein carbonylation and

decarbonylation." Antioxid Redox Signal 12(3): 393-404.

Xie, Z. J., Y. H. Wang, et al. (1990). "Studies on the specificity of the effects of oxygen

metabolites on cardiac sodium pump." J Mol Cell Cardiol 22(8): 911-20.

Yatime, L., M. Laursen, et al. "Structural insights into the high affinity binding of

cardiotonic steroids to the Na+,K+-ATPase." J Struct Biol 174(2): 296-306.

Zhang, C., S. Z. Imam, et al. (2002). "Peroxynitrite and the regulation of Na(+),K(+)-

ATPase activity by angiotensin II in the rat proximal tubule." Nitric Oxide 7(1):

30-5.

151 4.7 Figure Legends

Figure 4-1: A basal level of ROS is required in ouabain-induced activation of the Na/K-ATPase signaling.

LLC-PK1 cells were pretreated with NAC (10mM, 30min) or cobalt protoporphyrin (CoPP, 2µM) before ouabain (100nM, 15min) treatment.

Immunoprecipitation against total c-Src was performed to determine c-Src phosphorylation and its association with the Na/K-ATPase α1 subunit. Phospho-

ERK1/2 and total ERK1/2 were assessed with whole cell lysates. (a) A representative Western blot showing pretreatment with NAC prevented ouabain- induced pY418 c-Src and ERK1/2 phosphorylation. (b) and (c) Quantitative analysis of the effect of NAC pretreatment on ouabain-induced pY418 c-Src and ERK1/2 phosphorylation. The phosphorylation of c-Src and ERK1/2 was expressed as the ratio of phosphorylated c-Src (p-Src) and ERK1/2 (p-ERK) versus total c-Src (t-Src) and ERK1/2 (t-ERK), respectively. n=4. (d) The effect of NAC pretreatment on ouabain-stimulated association between total c-Src and the Na/K-ATPase α1 subunit. The association was expressed as ratio of the α1 versus total c-Src. n=4. **, p<0.01 vs con. #, p<0.01 vs ouabain treatment. (e) A representative Western blot showing that pretreatment with CoPP prevented ouabain-induced pY418 c-Src phosphorylation. (f) Quantitative analysis of the effect of CoPP pretreatment on ouabain-induced pY418 c-Src phosphorylation. n=4. **, p<0.01 vs con. #, p<0.01 vs ouabain treatment.

152 Figure 4-2: A basal level of ROS is required in ouabain-induced redistribution

of the Na/K-ATPase α1 subunit and NHE3, and inhibition of transcellular

22Na+ flux.

(a) A representative Western blot showing that pretreatment with NAC

(10mM, 30min) prevented ouabain (100nM, 1h) stimulated accumulation of the α1 subunit and NHE3 in early endosome (EE) fractions. Graph (b-d) show quantitative analysis of (a), n=4; ** p<0.01 vs control. (e) The effect of different concentrations

of NAC (30min) on ouabain (100nM, 15min)-induced pY418 c-Src phosphorylation,

n=3; ** p<0.01 vs control. (f) The effect of different concentrations of NAC

(30min) on ouabain (100nM, 1h)-induced inhibition of active transcellular 22Na+

flux from apical to basolateral compartment in LLC-PK1 monolayer grown on

Transwell® membrane support, n=3, ** p<0.01 vs control; * p<0.05 vs control.

Figure 4-3: Ouabain stimulated ROS generation and protein oxidative

modification.

Ouabain (100nM, 30min) stimulated ROS generation assessed by

Dihydroethidium (DHE) staining (a), formation of DMPO-nitrone adducts (b), and

protein carbonylation (c). Immunoprecipitation against DNP showed carbonylation modification of the α1 subunit, NHE3 and c-Src (d). Protein carbonylation might promote the accumulation of certain proteins in EE fractions (e).

153 Figure 4-4: Oxidative stress induced by GO caused similar effect as ouabain.

GO, like ouabain, activated c-Src (a) and ERK1/2 (b); n=3, ** p<0.01 vs

control; # p<0.05 vs ouabain or GO alone. GO (1 and 3 mU/ml, 1h) accumulated the α1 subunit and NHE3 in EE fractions in a dose-dependent manner (c-e); n=4, **

p<0.01 vs control. Unlike ouabain that only inhibits activate 22Na+ flux when added

to the basolateral aspect, GO (3 mU/ml, 1h) inhibits active 22Na+ flux in LLC-PK1

monolayer, effective on both apical and basolateral aspect (f); n=3, ** p<0.01 and*

p<0.05 vs control.

Figure 4-5: c-Src is required in ouabain-induced protein carbonylation, but

GO-mediated protein oxidation is partially dependent on c-Src.

A representative Western blot showing that pretreatment of c-Src inhibitor

PP2 (10µM, 30min) blocked phosphorylation of c-Src and ERK1/2 induced by GO

(3mU/ml, 1h) (a), abrogated ouabain (100nM, 1h)-mediated protein carbonylation and attenuated GO (3mU/ml, 1h)-induced protein oxidation (c). Ponceau S staining showing loading control. (b) Quantitative analysis of (a), n=4, ** p<0.01 vs control.

# p<0.05 vs GO. (d) A representative Western blot showing that ouabain (100nM,

1h) stimulated protein carbonylation in SYF+c-Src cells, but not in SYF cells. GO

mediated protein oxidation in both SYF+c-Src and SYF cells. (e) Quantitative

analysis of (d). n=4, ** p<0.01, # p<0.05 vs control.

Figure 4-6: Both Na/k-ATPase α1 subunit and caveolin-1 are necessary in

ouabain-mediated protein carbonylation.

154 Ouabain stimulated c-Src phosphorylation (a), an increase in superoxide

level (b) and protein carbonylation (c) in LLC-PK1 cells, but not in PY-17 cells and

C2-9 cells. Ponceau S staining showing loading control. n=3, ** p<0.01.

Figure 4-7: A ribbon diagram of the Na/K-ATPase with Pro222 and Thr224 highlighted (PDDB ID: 2ZXE).

155 4.8 Figures

Figure 4-1 a.

b.

# **

2

1 (relative)

p-Src/T-Src 0 n a o ua u C O NAC O +

NAC

156 Figure 4-1 c.

** # 2

1 (relative) p-ERK/t-ERK1/2 0

n C A Co Oua N

NAC+Oua

d.

# 2.5 **

2.0

1.5

1/T-Src 1.0 α (relative) 0.5

0.0

n C o ua A ua C O N +O

NAC

157 Figure 4-1 e.

f.

# ** 3

2

1 c-Src/ total c-Src

418 pY 0 con CoPP Oua CoPP+Oua

158 Figure 4-2 a.

b. # 2 **

1

1 inEE (relative) α 0

ua Con Oua NAC +O C NA

159 Figure 4-2 c.

3 # ** 2

(relative) 1 NHE3 EE in

0 a on AC C Oua N +Ou AC N

d.

2 # **

1 1 in LE 1 in α

(relative)

0

on C Oua NAC

NAC+Oua

160 Figure 4-2 e.

3 Control ** Oua ** 2

1

p-Src/T-Src (%) p-Src/T-Src 0

NAC= 0 1 5 10 mM

f. Control Oua 100 * ** Flux (%) + 50 Na 22

0

NAC 0 1 10 mM

161 Figure 4-3 a.

b.

162 Figure 4-3 c.

Ponceau S Staining

. d.

163 Figure 4-3 e.

Ponceau S Staining

164 Figure 4-4 a.

3 **, # 2 ** **

1 (relative%) P-Src/T-Src

0 l a u O O GO ontro a+G C u O

b.

165 Figure 4-4 c.

d.

4 ** ** 3 **

2 (relative)

p-Src/T-Src 1

0 n a 1 3 u O O Co O G G

166 Figure 4-4 e.

3 ** ** 2 ** 1 in1 EE α (relative) 1

0 n 1 Co Oua GO GO 3

f.

100 * ** ** Flux (%)

+ 50

Na 22 0 n ) ) o so so C a a (B (B a 3 O Ou G GO 3 (Apical)

167 Figure 4-5

a.

b. con PP2+GO PP2 GO 2.0 2.0 ** ** phospho-ERK1/2/T-ERK1/2 # # 1.5 1.5

1.0 1.0 c-Src/T--c-Src

418 0.5 0.5 PY

0.0 0.0 c-Src ERK1/2

168 Figure 4-5

c.

169 Figure 4-5

d.

e.

2.0 con oua # ** ** GO 1.5

1.0

0.5 Carbonyl contents

0.0 SYF + c-Src SYF

170 Figure 4-6

a.

Control 300 ** Oua 15min

200

100

(%) p-Src/T-Src 0 LLC-PK1 PY-17 C2-9

b.

300 ** con oua 100nM 1h 200

100

(%) DHE signal

0 LLC-PK1 PY-17 C2-9

171 Figure 4-6 c.

172 Figure 4-7

173 Chapter 5 – Summary and Conclusions

The overall aim of this dissertation is to improve our understanding of Na/K-

ATPase and NHE3 trafficking regulation and determine molecular mechanisms of

renal proximal tubular (RPT) sodium handling, which might contribute to the

impaired sodium excretion associated with hypertension.

In porcine RPT LLC-PK1 cells, we have shown that ouabain inhibits active

transepithelial 22Na+ transport (from apical to basolateral aspect) via protein trafficking regulation of the Na/K-ATPase and NHE3 (Liu, Kesiry et al. 2004; Cai

2008; Liu and Xie 2010), a process requiring ouabain-activated Na/K-ATPase signaling. This novel regulatory mechanism may contribute to CTS-induced renal sodium excretion. To investigate (1) if this regulatory mechanism is species- specific, (2) and if the internalized Na/K-ATPase and NHE3 is the endocytic recycling (reinsertion of endocytosed protein back to plasma membrane) in LLC-

PK1 cells, in manuscript 1 entitled “Ouabain-stimulated trafficking regulation of

the Na/K-ATPase and NHE3 in renal proximal tubule cells”, (1) three renal

proximal tubular cell lines (human HK-2, porcine LLC-PK1, and AAC-19

originated from LLC-PK1 cells in which the pig α1 was replaced by ouabain-

resistant rat α1) were employed to compare ouabain-induced regulation of the α1

subunit and NHE3 as well as transcellular 22Na+ transport, (2) LLC-PK1 cells were

used to assess endocytic recycling of NHE3 and Na/K-ATPase α1 subunit. Our

174 data indicate that the Na/K-ATPase is the functional receptor for ouabain-induced regulation of the Na/K-ATPase and NHE3 (and thus transcellular Na+ transport), as

we previously proposed (Liu and Xie 2010). This regulation is not species-specific,

but the species-specific α1 ouabain sensitivity may partially account for the species

differences observed in ouabain-induced natriuresis (Lloyd, Sandberg et al. 1992;

Yates and McDougall 1993; Loreaux, Kaul et al. 2008; Nesher, Dvela et al. 2009).

Specifically, the rodent α1 is far less sensitive response to ouabain than pig, dog, or

human α1. Higher concentrations of ouabain were required to activate Na/K-

ATPase signaling in rodents, compared to other species (Liu, Tian et al. 2000;

Aizman, Uhlen et al. 2001; Aydemir-Koksoy, Abramowitz et al. 2001; Haas, Wang et al. 2002; Liang, Cai et al. 2006). And the ouabain sensitivity of the α1 subunit influenced the ouabain-induced inhibition of 22Na+ flux as well as surface reduction of the α1 subunit and NHE3. Additionally, in ouabain-induced trafficking

regulation, endocytic recycling of internalized NHE3, but not the α1 subunit, was

inhibited by ouabain. The endocytosed α1 subunit, but not NHE3, is degraded through the LE- lysosome pathway.

In manuscript 2 entitled “Impairment of Na/K-ATPase signaling in renal proximal

tubule contributes to Dahl salt-sensitive hypertension”, we employed Sprague Dawley

rats, Dahl salt resistant (R) and sensitive (S) rats to investigate the role of Na/K-ATPase

signaling in renal sodium excretion and blood pressure regulation in vivo, further confirming our in vitro data: CTS function as natriuretic factors by CTS-induced redistribution of Na/K-ATPase and NHE3 and then RPT sodium transport regulation.

Our observations are as follows: (1) In Sprague Dawley rats, high salt diet activated c-Src

175 and induced redistribution of Na/K-ATPase and NHE3 in renal proximal tubules. (2) In

Dahl R rats, high salt diet significantly stimulated phosphorylation of c-Src and ERK1/2,

reduced Na/K-ATPase activity and NHE3 activity, and caused redistribution of Na/K-

ATPase and NHE3. In contrast, these adaptations were either much less effective or not seen in the Dahl S rats. (3) In primary culture of renal proximal tubule isolated from Dahl

S and R rats fed a low salt diet, ouabain induced Na/K-ATPase/c-Src signaling and

redistribution of Na/K-ATPase and NHE3 in the Dahl R rats, but not in the Dahl S rats.

These data indicate that impaired Na/K-ATPase signaling and consequent regulation of

Na/K-ATPase and NHE3 in renal proximal tubule may contribute to salt-induced

hypertension in the Dahl S rat.

Since there is no difference in the Na/K-ATPase α1 gene (Atp1a1) coding (Mokry and Cuppen 2008) and α1 sensitivity to ouabain (Nishi, Bertorello et al. 1993) between

Dahl salt-resistant and Dahl salt-sensitive rats, we propose that other factors must be present to affect the activation of Na/K-ATPase/c-Src signaling in the Dahl S rats.

It has been demonstrated that a high salt diet stimulates an increase in endogenous

CTS release and ROS generation. Our observations indicate that RPT sodium handling

via ouabain-mediated Na/K-ATPase/c-Src signaling appears to counterbalance blood

pressure rising caused by sodium retention, as shown in salt-sensitive hypertension (Liu,

Kesiry et al. 2004; Liu, Liang et al. 2005; Periyasamy, Liu et al. 2005; Oweis, Wu et al.

2006; Cai, Wu et al. 2008; Liu, Yan et al. 2011). Activation of Na/K-ATPase/c-Src

signaling, on the other hand, stimulates mitochondrial ROS production. In addition, c-Src

as initiating step of Na/K-ATPase signaling, is also redox-sensitive and required for ROS

generation (especially derived from NADPH oxidases) (Seshiah, Weber et al. 2002;

176 Touyz, Yao et al. 2003). Together with recent report that NADPH oxidase-derived ROS

production reduces renal proximal tubular sodium reabsorption (Panico, Luo et al. 2009),

we propose that there might be some cross-talk between Na/K-ATPase/c-Src signaling

and ROS generation in terms of renal sodium handling (Liu, Kennedy et al.)

In manuscript 3 entitled “Redox modulation of the Na/K-ATPase signaling and renal proximal tubular sodium handling”, we report that reactive oxygen species

(ROS) is critical in modulation of ouabain-Na/K-ATPase signaling and RPT ion transport based on the following observations: (1) Antioxidant NAC (N-Acetyl-L- cysteine) prevented ouabain-stimulated Na/K-ATPase signaling cascade, protein carbonylation, redistribution of the Na/K-ATPase and NHE3, and inhibition of transepithelial 22Na+ transport. (2) CoPP, HO-1 inducer (antioxidant enzyme),

attenuated ouabain stimulated-c-Src activation, which is an initiating step of Na/K-

ATPase signaling. (3) Protein carbonylation assay, spin-trap immunoassay, and DHE

staining all demonstrated that ouabain stimulated ROS generation and protein

oxidation. (4) c-Src is required in ouabain-induced protein carbonylation. (5)

Disruption of Na/K-ATPase/c-Src signaling attenuated ouabain-induced ROS

production. Interestingly, pretreatment with high doses but not low dose of NAC

attenuated ouabain-induced c-Src activation and blunted ouabain-inhibited active

transepithelial 22Na+ flux in LLC-PK1 cells, indicating that a basal physiological

range of ROS is necessary for initiation and amplification of ouabain-Na/K-ATPase

signaling and redistribution of Na/K-ATPase and NHE3. Furthermore, LC-MS/MS

analysis has identified that both ouabain and glucose oxidase caused direct

carbonylation of two amino acid residues in the actuator (A) domain of the Na/K-

177 ATPase α1 subunit. Taken together, our above findings unveil a novel mechanism

regulating RPT sodium handling via interaction of ROS with the Na/K-ATPase/c-

Src signaling pathway, which may influence salt retention related to volume

expansion that is responsible for hypertension. Moreover, direct protein

carbonylation in the actuator (A) domain of the Na/K-ATPase α1 subunit by ROS

might affect ouabain binding to the α1 subunit by induction of Na/K-ATPase

conformation change, favoring ouabain binding and c-Src activation, as well as

subsequently signaling cascade. However, the mechanisms remain largely to be

elucidated since the available data is limited (Garvin and Ortiz 2003; Schreck and

O'Connor 2011). Some pertinent questions remain to be resolved, such as how ROS

interacts with and influences the Na/K-ATPase/c-Src signaling cascade, as well as

whether ouabain-induced ROS generation acutely boosts the Na/K-ATPase signaling

by a positive feedback mechanism and chronically desensitizes the signaling cascade

by stimulating Na/K-ATPase/c-Src endocytosis.

Despite increasing evidence showing that antioxidants effectively lower

blood pressure in animal models, findings in human beings are less conclusive. A major clinical challenge is that routinely used antioxidants are ineffective in preventing or treating hypertension. The failure of antioxidant treatment prompts us to explore the underlying mechanisms that how ROS regulate blood pressure so that specific intervention can be used. We hope this dissertation and future in vitro and

in vivo studies on this topic would open up the possibility of translational clinical research and might improve the prognosis of hypertension and develop personalized patient management.

178 References for summary and conclusions

Aizman, O., P. Uhlen, et al. (2001). "Ouabain, a steroid hormone that signals with slow

calcium oscillations." Proc Natl Acad Sci U S A 98(23): 13420-4.

Aydemir-Koksoy, A., J. Abramowitz, et al. (2001). "Ouabain-induced signaling and

vascular smooth muscle cell proliferation." J Biol Chem 276(49): 46605-11.

Cai, H., L. Wu, et al. (2008). "Regulation of apical NHE3 trafficking by ouabain-induced

activation of the basolateral Na+-K+-ATPase receptor complex." Am J Physiol

Cell Physiol 294(2): C555-63.

Cai, H., Wu, L., Qu, W., Malhotra, D., Xie, Z., Shapiro, JI, and Liu, J. (2008).

"Regulation of Apical NHE3 Trafficking by Ouabain-Induced Activation of

Basolateral Na/K-ATPase Receptor Complex." Am J Physiol Cell Physiol 294(2):

C555-63.

Garvin, J. L. and P. A. Ortiz (2003). "The role of reactive oxygen species in the

regulation of tubular function." Acta Physiol Scand 179(3): 225-32.

Haas, M., H. Wang, et al. (2002). "Src-mediated inter-receptor cross-talk between the

Na+/K+-ATPase and the epidermal growth factor receptor relays the signal from

ouabain to mitogen-activated protein kinases." J Biol Chem 277(21): 18694-702.

Liang, M., T. Cai, et al. (2006). "Functional Characterization of Src-interacting Na/K-

ATPase Using RNA Interference Assay." J Biol Chem 281(28): 19709-19719.

Liu, J., D. J. Kennedy, et al. "Reactive Oxygen Species Modulation of Na/K-ATPase

Regulates Fibrosis and Renal Proximal Tubular Sodium Handling." Int J Nephrol

2012: 381320.

179 Liu, J., R. Kesiry, et al. (2004). "Ouabain induces endocytosis of plasmalemmal Na/K-

ATPase in LLC-PK1 cells by a clathrin-dependent mechanism." Kidney Int 66(1):

227-41.

Liu, J., M. Liang, et al. (2005). "Ouabain-induced endocytosis of the plasmalemmal

Na/K-ATPase in LLC-PK1 cells requires caveolin-1." Kidney Int 67(5): 1844-54.

Liu, J., J. Tian, et al. (2000). "Ouabain interaction with cardiac Na+/K+-ATPase initiates

signal cascades independent of changes in intracellular Na+ and Ca2+

concentrations." J Biol Chem 275(36): 27838-44.

Liu, J. and Z. J. Xie (2010). "The sodium pump and cardiotonic steroids-induced signal

transduction protein kinases and calcium-signaling microdomain in regulation of

transporter trafficking." Biochim Biophys Acta 1802(12): 1237-45.

Liu, J., Y. Yan, et al. (2011). "Impairment of Na/K-ATPase Signaling in Renal Proximal

Tubule Contributes to Dahl Salt-sensitive Hypertension." J Biol Chem 286(26):

22806-13.

Lloyd, M. A., S. M. Sandberg, et al. (1992). "Role of renal Na+,K(+)-ATPase in the

regulation of sodium excretion under normal conditions and in acute congestive

heart failure." Circulation 85(5): 1912-7.

Loreaux, E. L., B. Kaul, et al. (2008). "Ouabain-Sensitive alpha1 Na,K-ATPase enhances

natriuretic response to saline load." J Am Soc Nephrol 19(10): 1947-54.

Mokry, M. and E. Cuppen (2008). "The Atp1a1 gene from inbred Dahl salt sensitive rats

does not contain the A1079T missense transversion." Hypertension 51(4): 922-7.

Nesher, M., M. Dvela, et al. (2009). "Physiological roles of endogenous ouabain in

normal rats." Am J Physiol Heart Circ Physiol 297(6): H2026-34.

180 Nishi, A., A. M. Bertorello, et al. (1993). "Renal Na+,K(+)-ATPase in Dahl salt-sensitive

rats: K+ dependence, effect of cell environment and protein kinases." Acta

Physiol Scand 149(3): 377-84.

Oweis, S., L. Wu, et al. (2006). "Cardiac glycoside downregulates NHE3 activity and

expression in LLC-PK1 cells." Am J Physiol Renal Physiol 290(5): F997-1008.

Panico, C., Z. Luo, et al. (2009). "Renal proximal tubular reabsorption is reduced in adult

spontaneously hypertensive rats: roles of superoxide and Na+/H+ exchanger 3."

Hypertension 54(6): 1291-7.

Periyasamy, S. M., J. Liu, et al. (2005). "Salt loading induces redistribution of the

plasmalemmal Na/K-ATPase in proximal tubule cells." Kidney Int 67(5): 1868-77.

Schreck, C. and P. M. O'Connor (2011). "NAD(P)H oxidase and renal epithelial ion

transport." American Journal of Physiology - Regulatory, Integrative and

Comparative Physiology 300(5): R1023-R1029.

Seshiah, P. N., D. S. Weber, et al. (2002). "Angiotensin II stimulation of NAD(P)H

oxidase activity: upstream mediators." Circ Res 91(5): 406-13.

Touyz, R. M., G. Yao, et al. (2003). "c-Src induces phosphorylation and translocation of

p47phox: role in superoxide generation by angiotensin II in human vascular

smooth muscle cells." Arterioscler Thromb Vasc Biol 23(6): 981-7.

Yates, N. A. and J. G. McDougall (1993). "Effects of direct renal arterial infusion of

bufalin and ouabain in conscious sheep." Br J Pharmacol 108(3): 627-30.

181