INSIGHTS INTO THE RENAL PROTECTIVE MECHANISMS OF mRNA

BINDING HUR

DISSERTATION

Presented in Partial Fulfillment of the Requirements for

The Degree Doctor of Philosophy in the Graduate

School of The Ohio State University

By Mamata Singh Ohio State Biochemistry Program

The Ohio State University 2011

Dissertation Committee:

Professor Beth S. Lee, Advisor

Professor Richard W. Burry

Professor Arthur R. Strauch

Professor Ma Jiyan

ABSTRACT

Acute kidney injury is a common and serious problem in clinical medicine that can be roughly defined as an abrupt and sustained decrease in renal function resulting in failure of the kidneys to clear metabolic waste products.

Transient loss of blood flow to kidneys, or ischemia-reperfusion injury, is the most common cause of acute kidney injury, and results in damage to both renal tubule epithelia and endothelial cells, as well as generation of an inflammatory response. Under this type of stress, renal cells initiate programs to promote cell survival, although the nature of these adaptations is not well understood. Here we examine the function and expression of cell survival protein, HuR, in renal proximal tubule epithelia cells using an in vitro of ischemia-reperfusion injury.

HuR is a ubiquitously expressed protein that resides primarily in nuclei under normal growth conditions but translocates to the cytosol during stress, where it binds and stabilizes a select subset of mRNAs bearing adenine and uridine-rich sequences in their 3’ untranslated regions. Although HuR is generally regarded as an anti-apoptotic protein, recent studies have indicated that caspase cleavage of HuR may contribute to an amplified apoptotic response. To clarify

HuR’s role in renal stress, we knocked down or overexpressed it in proximal tubule cell lines that were subjected to ATP depletion, an in vitro procedure that mimics effects of ischemic injury to native kidneys. HuR was found to be protective against apoptosis that occurs when proximal tubule cells are ATP

ii depleted. Further, HuR was found to be necessary for full expression of the cell survival Bcl-2 and Hsp-70. These results strongly indicate an anti- apoptotic role for HuR in cultured proximal tubules cells, and suggest that its function in native proximal tubules may be protective against ischemic stress.

To understand what other cell survival signaling pathways might be induced by HuR, PCR array analysis was used to compare expression levels in

ATP-depleted cells with normal or suppressed levels of HuR. We found that loss of HuR caused decreased levels of mRNAs associated with the PI3-kinase/Akt signaling pathway that promotes cell survival. We found that HuR plays a central role in PI3K/Akt signaling, as not only did knockdown of HuR suppress many members of this pathway, but inhibition of PI3K also resulted in suppression of

HuR levels. Here, we show that in renal proximal tubule cells, HuR performs a central role in cell survival by amplifying Akt signaling in a positive feedback loop.

Key to this feedback loop is HuR-mediated stabilization of mRNA encoding

Grb10, an adaptor protein that enhances Akt activity by aiding in its transport to sites of activation. Under normal growth conditions, stimulation of Akt by HuR-

Grb10 interactions then activates NF-κB, which further enhances HuR expression at a transcriptional level. This study demonstrates a central role for

HuR in cell survival mechanisms and reveals how only modest changes in HuR levels below or above normal can be amplified, resulting in cell death, or alternately, cellular transformation.

The regulation of is a multi-level process that is essential to cellular processes. The data presented here show that the expression of HuR

iii during ATP depletion and recovery is upregulated by both transcriptional and

translational mechanisms. It was previously demonstrated that HuR mRNA is

expressed as two isoforms with vastly different 5’ untranslated regions and

different translatabilities. Here we have demonstrated that one of these mRNAs

is responsive to the BMP-7/Smad 1/5/8 signaling pathway, is more highly

expressed during recovery from ATP depletion, and is the only form capable of

being translated during ATP depletion. We also demonstrated that translation of the alternate form occurs during normal growth and recovery, and occurs through regulation by a post-transcriptional control element (PCE) present in its 5’ untranslated region. We further hypothesize that the differential translation of the

HuR isoforms may be due to their alternate targeting to polyribosomes or stress granules.

Stress granules are sites of aborted translation formed during various types of cell stress and recovery, including heat shock, arsenite poisoning, and UV- irradiation. Recruitment of specific mRNAs into stress granules regulates their stability and potential translation. We demonstrated that ATP-depleted epithelial cells form stress granules and activated their upstream kinases presumably for the purpose of regulating mRNA stability and translatability. During ATP depletion, some HuR mRNA is stored temporarily in stress granules. We hypothesize that this may be to avoid aberrant translation of HuR, which can lead

to tumorigenicity. The presence of a PCE imposes another level of regulation on

the translation of HuR and thus the translation of HuR is controlled during

normal, stressed, and recovered conditions.

iv

Dedicated to my Father, in his loving memory

v

ACKNOWLEDGEMENT

This dissertation would not have been possible without the guidance and

the help of several individuals who in one way or another contributed and

extended their valuable assistance in the preparation and completion of this

study.

First and the foremost, I am heartily thankful to my supervisor, Dr. Beth Lee,

whose encouragement, guidance and support from the initial to the final level

enabled me to develop a good understanding of the subject. I offer her my

gratitude, as she has supported me throughout my research with her patience

and knowledge along with providing excellent atmosphere for doing research.

Then, I would like to thank my committee members Dr. Richard Burry, Dr.

Arthur Strauch and Dr. Jiyan Ma for their precious suggestions and valuable comments that aided me in improving the quality of my research.

Additionally, I would like to thank my colleagues Dr. Selvi Jeyaraj, Dr. Dina

Ayupova, Dr. Brooke Mc Michael, and Suman Govindaraju for their constructive

criticism, help, and moral support.

I would also like to thank Alaina Martinez, a rotation student and a good

friend of mine, for all her help towards the completion of one of my projects.

vi Last but not the least, my parents, my husband, and my daughter for being patient with me, during all those years. In addition, I am thankful to the omnipresent divine spirit, for giving me the strength to finish this journey.

In the end, I offer my regards and gratitude to all of those who supported me in any respect during the completion of the project.

vii

VITA

July 20, 1974 …………………………………..Born-Banaras, India

June, 1997………………………………………B.S. Biochemistry

June, 1999………………………………………M tech in Biochemical Engineering IT BHU, India 2003-Present…………………………………...Graduate Teaching Assistant and Graduate Research Associate, The Ohio State University

PUBLICATIONS

1. Singh M, Sharma R, Banerjee UC. Biotechnological Applications of Cyclodextrins. J BiotechnologyAdvances 2002; 20:341-359.

2. Yi W Shao J, Zhu L, Li M, Singh M, Lu Y, Lin S, Li H, Ryu K, Shen J, Guo H, Yao Q, Bush CA, Wang PG. Escherichia coli O86 O-antigen biosynthetic gene cluster and stepwise enzymatic synthesis of human blood group B antigen tetrasaccharide. J Am Chem Soc. 2005 Feb 23; 127 (7):2040-1.

3. *Ayupova DA, *Singh M, Leonard EC, Basile DP, Lee BS. Expression of the RNA-stabilizing protein HuR in ischemia-reperfusion injury of rat kidney. Am J Physiol Renal Physiol. 2009 Jul; 97(1):F95-F105. (*denotes equal contribution)

4. Jeyaraj SC, Singh M, Ayupova DA, Govindaraju S, Lee BS. Transcriptional control of human antigen R (HuR) by bone morphogenetic protein (BMP). J Biol Chem. 2010; 2;285(7):4432-40.

FIELDS OF STUDY

MAJOR FIELD: BIOCHEMISTRY

viii

TABLE OF CONTENTS

ABSTRACT...... ii

ACKNOWLEDGMENTS...... vi

VITA...... viii

LIST OF FIGURES...... xiii

ABBREVIATIONS...... xv

CHAPTER 1

INTRODUCTION...... 1

1.1 Ischemia reperfusion Injury (IR)...... 1

1.2 Apoptosis: a mechanism of cell death...... 3

1.3 Ischemia Reperfusion Injury and Apoptosis….…...... 5

1.4 Cellular Models of Ischemic Injury………………………………………………..6

1.5 Differential Gene Expression as a Result of Ischemia Reperfusion Injury…..8

1.6 PI 3-Kinase Pathway, Apoptosis and Cell Survival……………………………..9

1.7 Mechanisms of Gene Regulation during Cell Stress……………………….…11

1.8 HuR and Gene Regulation…………………………………………….…….…...12

1.9 Role of mRNA Untranslated Regions in Gene Expression and mRNA Regulation……………………………………………………………………………...13

1.10 Stress Response-Stress Granules and Post-transcriptional Control Element (PCEs)…………………………………………………………………………………..14

1.10.1 Stress Granules ……………………………………………………….15

1.10.2 Post transcriptional elements (PCEs)…………………….…………17

ix

CHAPTER 2

MATERIALS AND METHODS……………………………………….……………....24

2.1 Cell Culture………………………………………………………………………..24

2.2 Apoptosis Assays…….. ……………………………………………………..…...24

2.3 Antibodies ………………………………………………………………………....25

2.4 Inhibitors…………………………………………………………………………...26

2.5 Western Analysis……………………………………………………………….…26

2.6 Transfection……………………………………………………………………..…27

2.7 RT-PCR and Immuno-RT-PCR. ………………………………………………..28

2.8 Immunocytochemistry……………………………………………………………29

2.9 PCR Arrays………………………………………………………………………...29

2.10 Plasmids………………………………………………………………………….30

2.11 In Situ Hybridization……………………………………………………………..30

2.12 Sucrose Density Gradients……………………………………………………..31

2.13 Gel Mobility Shift Assay………………………………………………………...32

2.14 Chromatin Immunoprecipitation (ChIP)………………………………………32

CHAPTER 3

HuR Promotes Cell Survival in ATP-depleted Renal Proximal Tubule Cells…..34

3.1 INTRODUCTION………………………………………………………………….34

3.2 RESULTS

3.2.1. Nature and Time Course of Cell Death in ATP-depleted LLC-PK1 Cells…………………………………………………………………………...... 37

x 3.2.2. Determination of Role of HuR in Cell Survival…………………… 38

3.2.3. Overexpression of HuR Promotes Cell Survival…………………..38

3.2.4. HuR Promotes Cell Survival by Inhibiting Both Intrinsic and Extrinsic Apoptosis……………………….………………………………………….…40

3.2.5. HuR Stabilizes Cell Survival mRNAs Bcl-2 and Hsp70 under Cell Stress………………………….………………………………………...... 41

3.3 CONCLUSION..………………………………………………………..………. .41

CHAPTER 4

HuR mediates anti-apoptotic effect by amplifying Akt signaling through a positive feedback loop, via Grb10……………………………………………………………50

4.1 INTRODUCTION………………………………………………………………...50

4.2 RESULTS

4.2.1 HuR Expression is Regulated by PI3K/Akt Activity in Proximal Tubule Cells…………………………………………………………………………...52

4.2.2. NF-κB is Critical for HuR Expression during Normal Growth…....54

4.2.3. Akt Activity but not Akt Expression is Regulated by HuR Levels in Proximal Tubule Cells……………………………………………………….55

4.2.4 Expression of Adaptor Protein Grb10 is Regulated by HuR Levels in Proximal Tubule Cells……………………………………………………….56

4.2.5. Grb10 Stimulates Akt Activation and HuR levels…………...... 58

4.3 CONCLUSION…………………………………………………………………...60

CHAPTER 5

Transcriptional and Translational control of HuR………………………………...68

5.1 INTRODUCTION…………………………..…………………………………….68

5.2 RESULTS xi 5.2.1 Gel Shift Assay and ChIP Confirms Binding of Smad 1/5/8 to HuR Promoter Elements……………………………………………...…………..69

5.2.2 Confirmation of the Presence of BMP-7 Receptors in LLC-

PK1cells………………………………………………………………………...71

5.2.3 Regulatory Mechanisms Mediating Translational Control of HuR...72

5.2.4 Translatability of Alternate Forms of HuR mRNA under Normal, Stressed, and recovered conditons…………………………………………72

5.2.5 Stress Granules………………………………………………………...73

5.2.6 Formation of Stress Granules during ATP Depletion and Recovery……………………………………………………………………….74

5.2.7 Effects of Cycloheximide and Puromycin on Stress Granule

Formation in LLC-PK1 Cells…………………………………………………75

5.2.8 Distribution of Stress Granule Markers during ATP Depletion and Recovery ………………………………………………………………………75

5.2.9 Increase in the Phosphorylation Level of eIF2-alpha during ATP Depletion……………………………………………………………………….77

5.2.10 PERK is Activated by ATP Depletion of Proximal Tubule Cells….78

5.2.11 Post-transcriptional Control Element (PCEs) and Translation of HuR……………………………………………………………………………..79

5.3 CONCLUSIONS…………………………………………………………………...81

CHAPTER 6

DISCUSSION………………………………………………………………………….92

6.1 Anti-apoptotic Role of HuR………………………………………………………92

6.2 HuR, PI3 Kinase/Akt Pathway, and Apoptosis………………………………...94

6.3 Transcriptional and Translational Control of HuR……………………………..97

BIBLIOGRAPHY……………………………………………………………………...100

xii

LIST OF FIGURES

Figure 1.1 Schematic of Intrinsic and Extrinsic Pathways of Apoptosis.………...20

Figure 1.2 Schematic of PI3K/Akt Signaling in Cell Survival and Growth……….21

Figure 1.3 The Role of eIF2α Phosphorylation in Global Translational Repression…………………………………………………………………...………...22

Figure 1.4 Formation of stress granules as a result of translational silencing….23

Figure 3.1 Time Course of Apoptosis in LLC-PK1 cells during ATP Depletion………………………………………………………………………………..44

Figure 3.2 Effects of HuR Knockdown on Apoptosis in ATP-depleted LLC-PK1 cells……...………………………………………………………………………………45

Figure 3.3 Distribution of Native and FLAG-tagged HuR in Stably Transfected

LLC-PK1 Cells……….………………………………………………………………...46

Figure 3.4 Effects of HuR Overexpression on Apoptosis in ATP-depleted LLC-

PK1 cells……………………..…………………………………………………………47

Figure 3.5 Effects of HuR Knockdown on Intrinsic and Extrinsic Pathways of Apoptosis……………………...... ……………………………………………………..48

Figure 3.6 Knockdown of HuR in ATP-depleted LLC-PK1 Cells Results in Suppression of Cell Survival Proteins…………………………..………………….49

Figure 4.1 HuR levels are regulated by PI3K/Akt signaling……………………...62

Figure 4.2 HuR levels are regulated by NF-κB signaling………………………..63

Figure 4.3 HuR regulates Akt activation and Grb10 expression…………………64

Figure 4.4 HuR regulates Akt activation and Grb10 expression…………………65

Figure 4.5 Knockdown of Grb10 inhibits Akt activation and HuR expression….66

xiii Figure 4.6 Schematic of HuR’s role in promoting a positive feedback loop of Akt signaling………………………………………………………………………………67

Figure 5.1 Smad 1/5/8 Binding Sites are Present in the HuR Promoter……….83

Figure 5.2 ATP Depletion and Recovery Induces Expression of ALK2………...84

Figure 5.3 Ribosomal Fractionation Indicates Differential Translation of HuR mRNAs during Normal Growth, ATP Depletion, and Recovery………………...85

Figure 5.4 ATP Depletion Induces Stress Granule Formation………………….86

Figure 5.5 Inhibitors of Translation Alter Stress Granule Formation in ATP- depleted LLCPK1 cells……………………………...………………………………..87

Figure 5.6 Neither eIF2-alpha nor a Large Ribosomal Subunit Co-Localize with

Stress Granules in LLC-PK1 Cells………………………………………………….88

Figure 5.7 Translation Initiation Factors, Small Ribosomal Proteins, and poly(A) mRNA Segregate to Stress Granules in LLC-PK1 Cells…………………….…..89

Figure 5.8 ATP Depletion Induces Phosphorylation of eIF2-alpha and PERK..90

Figure 5.9 HuR mRNA Contains a Post-transcriptional Control Element……....91

xiv

ABBREVIATIONS

AKI acute kidney injury

AMPK AMP-activated protein kinase

ARE AU rich element

ATN acute tubular necrosis

AUF1 AU rich binding factor-1

BMP bone morphogenic protein

DCP-1 decapping enzyme 1

ER endoplasmic reticulum

GAPDH glyceraldehyde-3-phosphate dehydrogenase

HuR human antigen R

HSP heat shock proteins

IRE iron response element

KIM-1 kidney injury molecule -1

MAP mitogen-activated protein

PABP poly A-binding protein

RHA RNA helicase A

TGF-β1 transforming β1

TIA-1 T-cell restricted intracellular antigen - 1

TIAR TIA-1 related

xv TTP tristetraprolin

SG stress granules

PCE post transcriptional control element

CX cycloheximide

PM puromycin

xvi

CHAPTER 1

Introduction

Excretion of waste and harmful chemicals and maintenance of fluid and solute balances are the major functions of the kidney. The kidney participates in whole-body homeostasis by regulating acid-base balance, electrolyte concentrations, extracellular fluid volume and regulation of blood pressure. The working unit of the kidney, the nephron, accomplishes this task through the process of plasma filtration along with reabsorption or secretion of solutes and water. Each kidney contains approximately 1,000,000 nephrons that contribute to regulation of blood and urine composition. Transient disruption of blood supply to the kidneys, or ischemia reperfusion injury, is the major cause of acute kidney injury (AKI) in both native and transplanted kidneys. The mortality of patients with

AKI remains high and unchanged in the modern era of critical care medicine.

1.1 Ischemia Reperfusion Injury (IR)

Restricted or transient interruption of the blood supply to tissues causes ischemic stress, which is a leading cause of the abnormalities resulting from myocardial infarction, cerebral ischemia, organ transplantation, and other short- lived traumas. Ischemic injury to tissues triggers a complex series of metabolic disturbances such as formation of oxygen radicals, changes in intracellular ion

1 concentrations, damage to the cytoskeleton, and alterations in gene expression

[Weinberg, 1991], often culminating in cell death. Ischemia is a state of tissue

oxygen deprivation accompanied by a reduced availability of the resulting metabolites [Bastin et al., 1987]. The absence of oxygen and nutrients from blood

creates a condition in which the restoration of circulation results in inflammation and oxidative damage through the induction of oxidative stress rather than

restoration of normal function. Restoration of blood flow to the ischemic tissue is

known as reperfusion. Despite the undeniable benefit of reperfusion of blood to

an ischemic tissue, reperfusion itself can educe a cascade of unfavorable

reactions that injure tissue [Becker, 2005]. Reperfusion injury has been known to

cause organ damage in the brain, heart, lungs, liver, kidneys and skeletal muscle

[Maxwell, 1997]. The major obstacle to reperfusion after an infarct or successful organ transplantation is the susceptibility of tissue to ischemia reperfusion (IR) injury. A key event in ischemia is the failure of cells to maintain adenosine triphosphate (ATP) levels, particularly proximal tubule cells, which are most susceptible to ischemic damage due to the high dependence on mitochondrial

ATP [Canfield et al., 1991]. Damage results from defective synthesis of ATP and degradation to adenosine. In addition, cellular ATP depletion results in mitochondrial dysfunction that initiates the translocation of Bax, a proapoptotic

Bcl-2 family member protein, from the cytosol to the outer mitochondrial membrane. This causes mitochondrial swelling and induces the efflux of cytochrome c into the cytosol where cytochrome c activates effector caspases and initiates apoptosis [Padnilam et al., 1999]. Although ischemic injury is

2 primarily due to oxygen-deprived cell death, reperfusion also produces a wide

array of inflammatory responses that causes local damage, which leads to

general insult [Gobe et al., 1999]. A growing body of evidence, primarily from

animal models of IR and preliminary human studies has revealed that

inflammatory mechanisms play a major role in the pathogenesis of IR [Maxwell et

al., 1997].

1.2 Apoptosis: a mechanism of cell death

The word apoptosis is derived from Greek origins and means falling off or

senescence. Unlike the traumatic destruction of necrosis, apoptosis refers to a

regulated or programmed cell death, which is also called death by suicide [Kerr et al., 1972]. Apoptosis may play a positive role in organismal biology. During development, excess cells undergo programmed cell death to form final tissue structures. Alternately, dysregulated apoptosis results in negative consequences.

Excessive apoptosis causes atrophy of tissues, such as in ischemic damage, whereas insufficient levels of apoptosis may result in uncontrolled cell proliferation, such as cancer.

Morphological changes in apoptotic cells include cell shrinkage, deformation

and loose contact to neighboring cells, chromatin condensation, plasma membrane blebbing, and finally fragmentation into compact membrane-enclosed

structures, called “apoptotic bodies”. The apoptotic bodies can be engulfed by

macrophages and thus may be removed from the tissue without causing an

inflammatory response. Apoptosis leads to activation of proteolytic enzymes,

3 which eventually mediate the cleavage of DNA into oligonucleosomal fragments as well as the cleavage of a multitude of specific protein substrates which usually determine the integrity and shape of the cytoplasm or organelles [Saraste and

Pulkki, 2000]. Apoptosis is opposite to the necrotic mode of cell death in which cells suffer a major insult, resulting in a loss of membrane integrity, swelling, and disruption. During necrosis, cellular contents are released uncontrolled into the immediate environment which results in damage of surrounding cells and a strong inflammatory response in the corresponding tissue [Leist, 2001].

Apoptosis occurs through two main signaling cascades, the extrinsic and intrinsic apoptotic pathways. Any stimulus that causes oxidative stress, mitochondrial disturbances and DNA damage induces the intrinsic pathway. For example, cancer therapeutic agents, hypoxia, and ionizing irradiation can trigger the intrinsic pathway of apoptosis. When the mitochondrion is damaged, the outer membrane becomes permeable and facilitates cytochrome c release into cytoplasm. Further, cytochrome c binds to the caspase adaptor, Apaf-1

(apoptotic protease-activating factor-1), to form the apoptosome complex that consequently triggers the apoptotic cascade by activating procaspase 9. The newly activated caspase 9 activates many downstream effector caspases including caspases 3, 6 and 7, leading to DNA fragmentation and cell death

[Earnshaw, 1999]. On the other hand, the extrinsic apoptotic pathway is induced by ligand binding to death receptors. The major ligand-death receptor systems include tumor necrosis factor (TNF) with tumor necrosis factor receptor 1

(TNFR1), Fas ligand with Fas, and TRAIL with TRAIL receptors. Binding of the

4 receptors with ligands induces receptor oligomerization and recruitment of death

signal adaptor proteins. These form a complex termed DISC (death-inducing

signaling complex), which binds to initiator caspases 8 and 10. Consequently, the

caspase cascade is triggered to activate the caspases 3, 7 and 9, leading to

apoptotic events [Ashkenazi, 2002]. There exists extensive crosstalk between the

intrinsic and extrinsic apoptotic pathways, as activation of the caspase cascade

is the crucial component in the death process in either pathway. An overview of

these cascades is represented diagrammatically in Figure 1.1.

1.3 Ischemia Reperfusion Injury and Apoptosis

Renal tubular epithelial cell injury from ischemia was traditionally regarded

as a result of necrotic form of cell death. However, one of the first studies to describe apoptosis in ischemic acute renal failure was by Schumer et al. in 1991, in which DNA fragmentation in the kidney cortex was detected 12 hours after reperfusion. Similarly, several studies using immunohistochemical techniques reported the typical ladder pattern of DNA after subjecting kidneys to IR.

Additional evidence for the importance of apoptosis in IR has been provided by recent studies by Kelly et al. in 2003, who demonstrated the importance of guanosine triphosphate (GTP) depletion in apoptosis (versus ATP in necrosis) and the ability of guanosine and p53 inhibitors to protect against apoptosis and renal dysfunction.

In kidneys, the extrinsic pathway of apoptosis is initiated by activation of cell

death receptors for Fas and tumor necrosis factor-α (TNF-α) leading to activation

5 of procaspase-8, which, in turn, cleaves and activates procaspases-3. The

mitochondrial-dependent pathway is triggered by cytochrome c release from the

mitochondria [Castenada et al., 2003]. Cytochrome c binds to Apaf-1, leads to

caspase-9 activation. Differential regulation of caspases in kidneys subjected to

I/R injury have been shown [Devarajan, 2006]. The cleavage of caspase-1 and

caspase-3 is documented during reperfusion indicating activation of these

enzymes. In addition, a rat model of IR indicated that prolonged ischemia

induced pro-apoptotic mechanisms, including increases in the Bax/Bcl-2 ratio,

caspase-3 expression, poly [adenosine triphosphate (ADP)-ribose] polymerase

(PARP) cleavage, DNA fragmentation, and apoptotic cell number in renal

proximal and distal tubules [Wei et al., 2004].

In living cells, mitochondrial changes are predominantly prevented by

antiapoptotic members of the Bcl-2 family of proteins. It has been shown that the

activation of caspase-3 during hypoxia or ATP depletion is accompanied by the

translocation of Bcl-2 family member Bax from the cytosol to the mitochondria

and the release of cytochrome c from the mitochondria to the cytosol [Schwarz et

al., 2002]. However, transfecting the cells with Bcl-2 provided protection against

hypoxia-induced injury. Further, recent studies have recognized

phosphatidylinositol 3 (PI3) kinase/Akt phosphorylation as one of the signaling

pathways that blocks apoptosis and promotes cell survival in response to diverse apoptotic stimuli in different cell types [Devarajan, 2006].

1.4 Cellular Models of Ischemic Injury

6 Renal epithelial cell lines provide a convenient system for studying many

processes that occur in the kidney. Cell lines from many species exist that

represent the distal and proximal tubular epithelia. Because proximal tubules are

most susceptible to injury during renal ischemia, proximal tubule cell lines have

been well established as cellular models of renal ischemia. In particular, the LLC-

PK1 porcine proximal tubule cell line has been well studied in this regard. LLC-

PK1 cells were originally isolated from the kidney cortex of Hampshire pigs.

These cells maintain many characteristics of proximal tubule epithelia. Once

confluent, these cells maintain polarized basal and apical membranes, form tight

junctions between cells, form a brush border and continue cell transport systems

specific to this cell type [Amsler and Cook, 1982; Mullin, 1980]. The LLC-PK1 cell

line has also been well established as a model for studying stress to proximal

tubule epithelia. Depleting these cells of ATP results in alterations similar to the

ischemic kidney including activation of heat shock proteins [Van SK et al., 2003],

loss of cellular polarity due to disruption of the actin cytoskeleton [Canfield,

1991], and activation of protein kinases [Park KM et al., 2002].

Similarly, HK-2 cells are proven to have a potential usefulness as a tool for

studying injury and repair. HK-2 is an immortalized proximal tubule epithelial cell

line from normal adult human kidney. Phenotypically, the cell line HK-2 has the

same characteristics of adult normal tubular cells [Ryan et al., 1994]. In

particular, HK-2 cells maintain the brush border typical enzymatic activities (acid and alkaline phosphatase, leucine aminopeptidase, gamma

glutamyltranspeptidase). Furthermore, HK-2 cells retain functional characteristics

7 of proximal tubular epithelium (Na+ dependent/phlorizin sensitive sugar transport; adenylate cyclase responsiveness to parathyroid, but not to antidiuretic, hormone). Therefore, this cell line is used as a valuable tool for the study of physiological and pathophysiological human renal tubule, as well as the mechanisms of damage and repair at the level of tubular cell [Nadia et al., 2008].

1.5 Differential Gene Expression as a Result of Ischemia Reperfusion Injury

Global gene expression is altered during ischemia reperfusion injury. cDNA microarray has been a powerful tool to detect molecular changes during renal IR.

Altered gene expression includes changes in the levels of transcription factors, growth factors, signal transduction molecules, and apoptotic factors [Yoshida et al., 2002]. Upregulation of several transcription factors, such as AP-1, and Zf9 kruppel-like transcription factor has been noted. IR-regulated growth factors include heparin-binding epidermal growth factor (HB-EGF), insulin-like growth factor 1 (IGF-1), transforming growth factor-β1 (TGF- β1), and cell cycle proteins like p21 [Devarajan et al., 2003]. Signal transduction proteins that are induced during ischemia reperfusion events include transforming growth factor-1, interleukin 2 receptor, gamma interleukin 4 receptor, growth factor receptor bound (Grb) 2, Ras-like guanosine triphosphate binding protein, CD151, and endothelial nitric oxide synthase 3. In addition, some cytoskeletal proteins like claudin 7, cadherin 5 alpha, smoothelin, palladin, talin, vinculin, , tropomyosin 4, filamin, and β-tubulin also demonstrate changes in their gene profile [Supavakin et al., 2000].

8 Apoptotic factors such as Fadd, Bad, Daxx, Bax and p53 were found to be upregulated as suggested by micro-array analysis in a rat kidney ischemia- reperfusion model [Harris, 1997]. Both the death receptor-dependent (Fadd-

Daxx) and mitochondrial (Bad-Bak) pathways were found to be activated. This indicates that apoptosis is an important mechanism for the early loss of tubule cells following ischemia/reperfusion injury. In addition, activation of heat shock proteins 27, 72 and 70 as well as protein kinases JNK, MAPK kinase, and p38 has been observed during stress [Devarajan et al., 2003].

Understanding these changes in gene expression not only provides insight

into the pathogenesis of ischemia/reperfusion injury, but also can potentially

provide novel biomarkers and potential therapeutic targets for the treatment of

ischemic injury.

1.6 PI 3-Kinase Pathway, Apoptosis and Cell Survival

Signaling by TNF and related polypeptides is an active process in which

stimulation of cell death receptors induces apoptosis. Other signaling pathways

act in the opposite direction to promote cell survival by inhibiting apoptosis.

These pro-survival pathways control the fate of a wide variety of cellular survival

processes. One of the major intracellular signaling pathways responsible for

promoting cell survival is initiated by the enzyme PI3-kinase, which is activated

by either receptor tyrosine kinases or G protein-coupled receptors [Song et al.,

2005]. PI3-kinase phosphorylates the membrane phospholipid PIP2 to form

PIP3, which activates the protein serine/threonine kinase Akt. PIP3 recruits the

9 protein kinase Akt to the plasma membrane where it is activated as a result of

phosphorylation by PDK. Akt then phosphorylates a number of proteins that

contribute to cell survival. Akt (also known as protein kinase B) was originally

identified as the cellular homologue of the transforming oncogene of the AKT8

retrovirus. Several pathways downstream of PI-3/Akt phosphorylation have been

proposed for cell survival [Datta et al. 1999]. One of the well-studied molecules

that mediate cell survival by Akt phosphorylation is the proapoptotic Bcl-2 family

member Bad. Bad has the ability to directly interact and bind to antiapoptotic Bcl-

2 and Bcl-XL and block their survival function. Phosphorylated Akt can directly

phosphorylate Bad both in vitro and in vivo, and makes Bad incapable of binding

to Bcl-XL, thus restoring its antiapoptotic function. Sequestering phosphorylated

Bad in the cytosol also makes it unavailable to bind to Bcl-2, thus preventing it from damaging the mitochondria [Orike et al., 2001]. Akt can also phosphorylate human caspase-9, resulting in reduction of caspase-9 activity. Further, Akt

activates transcription factor NF-κB via regulating IκB kinase (IKK), resulting in

transcription of pro-survival [Datta et al., 1999].

In addition to these direct effects on components of the cell death

machinery, Akt phosphorylates transcription factors that regulate cell survival and

another protein kinase (GSK-3) that affects apoptosis as well as regulating cell

metabolism and protein synthesis. The PI3-kinase/Akt pathway thus regulates

cell survival through phosphorylation of a variety of downstream targets, extracellular growth factors or cell-cell interactions [Dancey, 2004]. A schematic

10 depicting the central role of PI 3-kinase/Akt signaling in cell survival is shown in

Figure 1.2.

PI3-kinase and Akt are activated after renal ischemia/reperfusion, leading to phosphorylation of FKHR and FKHRL1, which may affect epithelial cell fate in acute renal failure [Andreucci et al., 2003]. Inhibiting PI3-kinase is known to impose deleterious effects on serum blood urea nitrogen level and renal functions in mice [Xie et al., 2003]. PI3-kinase is also reported to activate PKC, which is involved in preconditioning, suggestive of its role in cell survival. Akt has been suggested to promote cell survival by intervening in the apoptosis cascade even before cytochrome c release and caspase activation [Kennedy et al., 1999].

In addition, PI3-kinase/akt activation leads to the activation of MEK/ERK survival signaling pathways through the reactive oxygen species-dependent

EGFR/Ras/Raf cascade [Kwon et al., 2006]. Activation of these kinases may be involved in the repair process during ischemia/reperfusion. Thus, PI3-K/Akt signaling pathway plays an important role in regulating the repair following renal

IR.

1.7 Mechanisms of Gene Regulation during Cell Stress

Following stress injury, cells undergo an adaptive response to alter gene

expression. Gene expression is under the coordinated control of mRNA

transcription, mRNA translation, and degradation. It follows that, since turnover of

certain mRNAs is an important form of regulation in mammalian cells, then

stabilization of certain mRNAs is vital for cell survival. Stable mRNAs permit a

11 longer translational window for highly expressed genes or a delayed expression

window when they are stably stored and expressed in a temporal manner.

Whereas the roles of transcription and degradation rates have been well studied,

the role of mRNA stability is now emerging as an important step in regulation.

There are many factors involved in determining mRNA stability. These include primary and secondary structure, translation rate, and intracellular location.

1.8 HuR and Gene Regulation

HuR is a widely expressed nucleocytoplasmic shuttling protein possessing

three well-conserved RNA recognition motifs [Peng et al., 1998]. Under normal

growth conditions, the majority of HuR is maintained in the nucleus but moves to

a more cytoplasmic distribution under stressed conditions [Jeyaraj et al., 2006].

In the cytoplasm, HuR aids in regulating stability and translatability of ARE- containing mRNAs by binding and stabilizing its cognate mRNAs for future translation [Fan et al., 1998]. HuR binds to specific mRNAs that contain adenosine-uridine-rich elements (AREs) in their 3’ untranslated regions, and stabilizes them during stress [Lal et al., 2003].

HuR, as one of the regulators of mRNA stability in renal epithelial cells, is

found to be redistributed and up-regulated at multiple levels during energy

depletion and recovery. By binding to mRNAs containing specific AREs, it

contributes to the stability of hundreds to thousands of distinct mRNA transcripts.

These proteins belong to a several major categories, including modulators of cell

proliferation and differentiation, inflammation, cell survival and apoptosis. It is a

12 stress response protein and is known to play an important role in cell survival, as

its loss in vitro or in vivo results in heightened apoptosis [Wang et al., 2000]. The

expression of HuR as a result is tightly regulated, as its over expression is

associated with oncogenicity in multiple tissues including renal carcinoma cell

lines [Denkert et al., 2004].

HuR has been studied under several forms of stress in cell culture models;

however, its potential role in organ systems has not been well explored. Because

HuR appears to play a role in stabilizing numerous transcripts during cellular

stress and apoptosis [Wang et al., 2000], understanding the regulation of this

protein will give important insight into general mechanisms of survival during

environmental insults such as ischemic injury.

1.9 Role of mRNA Untranslated Regions in Gene Expression and mRNA

Regulation

As described above, HuR binds to cognate mRNAs through specific

sequences in their 3’ untranslated regions (UTRs). Gene expression is finely

regulated at the post-transcriptional level by untranslated regions both 5’ and 3’

to the mRNA coding region [Chen et al., 1995]. Features of UTRs that control

mRNA translation, degradation, and localization include stem-loop structures, upstream initiation codons and open reading frames, internal ribosome entry sites and various cis-acting elements that are bound by RNA-binding proteins.

UTRs are known to play crucial roles in transport of mRNAs into and out of the nucleus, translation efficiency [Velden and Thomas, 1999], subcellular

13 localization [Jansen, 2001] and stability [Bashirullah et al., 2001]. Regulation by

UTRs is mediated in several ways. Nucleotide patterns or motifs that are the

target of trans-acting RNA binding proteins located in 5' UTRs and 3' UTRs can interact with specific RNA-binding proteins. Secondary structures in 5' UTRs are also important in the regulation of translation. Structural features of the 5' UTR have a major role in the control of mRNA translation [Sweeny, 1996]. The importance of UTRs in regulating gene expression is underlined by the finding that mutations that alter the UTR can lead to serious pathology [Conne et al.,

2000].

1.10 Stress Response-Stress Granules and Post-transcriptional Control

Element (PCEs)

In response to stressful environmental conditions, cells educe a series of

adaptive changes known as the stress response. Central to the stress response

is translational repression of all but a minority of stress- induced proteins. In most

cases, this general translational repression is due to hyperphosphorylation of the

translation initiation factor elF2α, a subunit of eukaryotic initiation factor elF2.

eIF2 activity is regulated by a mechanism involving both guanine nucleotide

exchange and phosphorylation. Phosphorylation takes place at the α-subunit, which is a target for four different kinases that phosphorylate serine 51. Those

kinases act as a result of stress such as amino acid deprivation (GCN2), ER

stress (PERK), the presence of dsRNA (PKR) or hemoglobin deficiency (HRI).

During translation initiation, elF2α forms a complex with GTP and the initiator

14 methionyl-tRNA, and this complex binds to the small ribosomal subunit to initiate

protein synthesis. One subunit of elF2α, eIF2B, is a guanine nucleotide

exchange factor that regulates the activity of the elF2 complex. Exchange of

GDP for GTP is prevented by phosphorylation of elF2α, thereby reducing the

availability of the elF2-GTP-met-tRNA complex, as a result translation cannot

proceed [Kedersha et al., 2002]. The relationships among members of the eIF2

complex and their roles in translation initiation are indicated in Figure 1.3. Along

with this general translational repression under stress, other regulatory pathways exist that shuttle the mRNA templates of selected proteins into active polysomes.

1.10.1 Stress Granules

Under conditions of stress, cells form discrete cytoplasmic foci called stress

granules (SGs). These have been identified in plant, yeast, and animal cells and

are characterized as sites of stalled translation initiation [Kedersha and

Anderson, 2002]. The assembly of SGs is triggered by phosphorylation of elF2α, which effectively prevents formation of the eIF2-GTP-Met-tRNAi ternary complex

[Kimball et al, 1999]. The RNA binding proteins TIA-1 and TIAR act downstream of the phosphorylation of eIF2 and recruit these untranslated mRNAs to SGs

[Kedersha et al., 2002]. An overview of stress granule formation is depicted in

Figure 1.4. The purpose of stress granules may be to protect RNAs from harmful conditions, to promote cell survival, and to function as a decision point for untranslated mRNAs [Kimball et al., 2003]. These mRNAs subsequently can go

15 down one of three paths: further storage, degradation, or re-initiation of translation.

Stress granules are composed of silent pre-initiation complexes containing small ribosomal proteins, as well as numerous mRNA-binding proteins such as

TIA-1, TIAR, poly-A binding protein (PABP), HuR, tristetraprolin, and several translation initiation factors [Kedersha et al., 1999]. These granules appear to act as mRNA storage sites, regulated in part by mRNA binding proteins (such as

HuR and TIAR) that mediate the stability of specific transcripts. Nearly all of the mRNA in the cell is recruited to SGs during stress [Kimball et al., 2003], however, specific transcripts that are critical for cell survival during stress (e.g. Hsp70) are excluded from the stress granules and are shuttled into active polysomes.

Considering total cellular mRNA, these critical mRNAs are in minority. For example, viral infection causes cell stress, but only about 3 - 5% of cellular mRNAs become associated with polysomes during infection [Kedersha et al.,

2002].

Only certain types of environmental insults such as heat shock, osmotic shock, UV irradiation, oxidative stress, and energy depletion lead to the formation of stress granules, whereas other forms of insult, including serum starvation, inhibition of transcription, microtubule or microfilament depolymerization, and inflammatory cytokines, do not result in formation of SGs [Kedersha et al., 1999].

It has also been observed that energy-depleted cells are able to form stress granules in the absence of eIF2α phosphorylation and it has been suggested that the lack of GTP in the system inhibits formation of the eIF2-GTP-met-tRNA

16 complex, which is required for translation initiation [Kedersha et al., 2002].

However, this contradicts the notion that elF2α phosphorylation is necessary for

SG formation [Kimball et al., 2003].

1.10.2 Post-transcriptional Elements (PCEs)

Following viral infection or disease, efficient regulation of translation is important in response to adapt to environmental changes. Translational initiation starts with the formation of multi-protein complex eukaryotic initiation factor eIF-

4A, recruitment of the small ribosomal subunit, scanning for the initiation codon and subsequent recruitment of the large ribosomal subunit. Like all other steps of translation, this step is tightly regulated. Transcripts containing a short (<100 nt), relatively unstructured 5' UTR are generally good candidates for efficient ribosome scanning. Conversely, transcripts that contain a longer and highly structured (i.e. G+C-rich) 5' UTR are less efficiently scanned. The structural features of 5' UTR, and the type of ribonucleoprotein complex (RNP) binding, determines the ribosome scanning and the efficiency of translation initiation.

Retroviral proteins are known for their long transcripts, containing highly structured 5’ UTRs. Long 5’ UTRs having inhibitory characteristics prompt for alternative mechanisms for translation. Translation through and internal ribosome entry site (IRES) is an alternative to the traditional cap dependent translation.

IRESs recruit ribosomal initiation complexes directly to the translation start codon by avoiding the scanning procedure through a highly structured 5’ UTR.

Recently, a novel highly structured RNA element in the 5’ UTR of selected

17 retroviruses and a cellular mRNA has been revealed. This element, called the

posttranscriptional control element (PCE), facilitates, rather than inhibits, cap

dependent translation.

PCEs are redundant stem-loop RNA structures that were initially identified

in the 5' UTR of avian spleen necrosis virus (SNV) [Roberts et al., 2005] and the

later in retroviruses including Mason-Pfizer (MPMV) [Hull at al., 2002], human

foamy virus (HFV) [Russell et al., 2001], reticuloendotheliosis virus strain A

(REV-A), human T-cell leukemia virus type 1 (HTLV-1), feline leukemia virus

(FeLV), bovine leukemia virus (BLV) [Boris-Lawrie et al., 1995] and HIV-1 [Zapp

et al., 1989]. PCEs are highly structured 5’ UTR sequences that facilitate cap

dependent translation. A protein called RNA helicase A (RHA) is known to

associate with the PCE structures [Hartman et al., 2006]. RHA is a

multifunctional DEIH box helicase and RNA binding protein, and deregulation of

RHA has been associated with various cancers and autoimmune disease

[Tettweiler et al., 2006]. RHA operates as a selective translation control switch

and facilitates cap-dependent translation. RHA interacts with PCE-containing

mRNAs in the nucleus and cytoplasm, and contributes to RNA/RNP remodeling.

Through its helicase and/or RNPase activity, RHA assists the melting of the

complex structure of the PCE to enable its efficient association with polysomes.

Upon RHA knock down, PCE-containing mRNAs accumulate in the cytoplasm,

though they are translationally silent and are sequestered in RNA storage

granules. Similarly, non-functional PCE mutant RNAs accumulate in the

18 cytoplasm in stress granules as these transcripts lack efficient interaction with

RHA and are poorly translated [Ranji et al., 2010].

1.11. Overview

The following studies will be an exploration of the role of HuR in protecting renal tubule cells against apoptosis caused by ischemia-reperfusion injury and

ATP depletion, as well as the transcriptional and translational mechanisms by which HuR itself is regulated. These studies have been designed to illuminate key aspects of the mechanisms behind cell survival and repair during acute kidney injury, and to uncover the role of HuR in this process.

19

Figure 1.1. Schematic of Intrinsic and Extrinsic Pathways of Apoptosis. The figure is adapted from Cellsignal.com.

20

Figure 1.2. Schematic of PI3K/Akt Signaling in Cell Survival and Growth. Figure is adapted from sarcomahelp.org/research

21

Figure 1.3. The Role of eIF2α Phosphorylation in Global Translational Repression. During translation initiation, elF2α forms a complex with GTP and the initiator methionyl-tRNA, and this complex binds to the small ribosomal subunit to initiate protein synthesis. One subunit of elF2α, eIF2B, is a guanine nucleotide exchange factor that regulates the activity of the elF2 complex. Exchange of GDP for GTP is prevented by phosphorylation of elF2α (by various kinases like GCN2, PKR, HRI, PERK), thereby reducing the availability of the elF2-GTP-met-tRNA complex, as a result translation cannot proceed. Figure is adapted from deverlab.nichd.nih.gov.

22

Figure 1.4. Formation of stress granules as a result of translational silencing. The schematic in (A) illustrates normal translation initiation. In (B), phosphorylation of eIF2α by various protein kinases, including PKR, results in aborted translation initiation, recruitment of mRNA binding proteins such as TIA- 1, and failure of large (60S) ribosomal subunits to join the 40S complex. Figure adapted from Cell Stress Chaperones 2002; 7(2):213-221.

23

CHAPTER 2

Materials and Methods

2.1 Cell Culture

LLC-PK1 or HK-2 cells (American Type Culture Collection, Manassas, VA)

were cultured in Dulbecco’s modified Eagle’s medium containing

penicillin/streptomycin supplemented with fetal bovine serum (10%) at 37°C in

5% CO2. For depletion of ATP from these cells, cultures were first grown to

confluency, then medium was replenished and cultures were incubated

overnight. The cells were then rinsed twice with phosphate-buffered saline and

the culture medium was replaced with pre-warmed Dulbecco’s modified Eagle’s medium base supplemented with L-glucose, sodium bicarbonate, and 0.1 μM

antimycin A for 0–6 hours. In some experiments, ATP-depleted cells were

allowed to recover by returning the cells to normal growth medium.

2.2 Apoptosis Assays

For caspase assays of siRNA treated cells, LLC-PK1 or HK-2 cells were

seeded in 96-well plates, grown to confluence, and transfected with targeted or

control siRNAs. Cells were subjected to ATP depletion 48 hours post-transfection and assayed for caspase 3/7 activity using the Caspase Glo 3/7 Assay

24 (Promega, Madison, WI). HuR-overexpressing cells or controls were seeded at

12 x 103/well, grown to confluence, subjected to ATP depletion, and assayed for caspase 3/7 activity using the same kit. Each treatment was performed in triplicate, and three such independent experiments were performed. For enumeration of apoptotic nuclei, siRNA-treated, HuR-overexpressing, or control cells were cultured on glass coverslips in 24-well plates. Following ATP depletion, cells were permeabilized, fixed with 4% paraformaldehyde and stained with Hoechst stain (1 μg/μl). Samples were examined under a Nikon Eclipse 80i

epifluorescent microscope. Condensed or fragmented nuclei were scored as

apoptotic, and three similar fields of cells were enumerated for each time point.

To test the role of PI3 kinase signaling, HK-2 cells were treated with 50 µM LY-

294002 or vehicle for 16-18 hours prior to ATP depletion and subsequent

caspase assays.

2.3 Antibodies

A mouse monoclonal antibody (3A2) against HuR, a rabbit polyclonal against

Bcl-2, and rabbit polyclonal antibodies recognizing mouse Grb10 and Smad1/5/8

were purchased from Santa Cruz Biotechnology (Santa Cruz, CA). A mouse

monoclonal against Hsp70, TIAR and eIF3 was purchased from BD Biosciences

(San Jose, CA). Antibodies recognizing ALK2 and β-actin were purchased from

Abcam (Cambridge, MA). Rabbit antibodies against total Akt, phospho-Akt, eIF2

alpha, phospho-eIF2 alpha, PERK, phospho-PERK, and S6, were obtained from

Cell Signaling Technology (Danvers, MA). A rabbit antibody that detects human

25 Grb10 also was purchased from Technology. Secondary

antibodies coupled to horseradish peroxidase were purchased from

Amersham/GE Healthcare (Piscataway, NJ). Secondary fluorescent antibodies like Alexa 488-or Alexa 568-conjugated goat anti-mouse antibodies were bought

from Molecular Probes (Eugene, OR).

2.4 Inhibitors

The PI3 kinase inhibitor LY-294002 (Cell Signaling Technology) was added to cultures, where appropriate, at a concentration of 50 μM for 16-18 hours prior

to ATP depletion experiments. The NF-κB inhibitor BAY11-7082 (Sigma-Aldrich,

St. Louis, MO) was used at a concentration of 2 µM. Translation inhibitors

cycloheximide and puromycin were obtained from Sigma-Aldrich and used at

concentrations of 100 µg/ml and 10 µg/ml, respectively.

2.5 Western Analysis

Cell lysates were prepared by incubating cells in MPER with protease and

phosphatase inhibitor cocktails (Pierce, Rockford, IL). Cell lysates were

subjected to centrifugation at 10,000 rpm for 5 minutes for the removal of cell

debris. The supernatant protein content was determined using a BCA protein

assay kit (Pierce). For Western blot analysis, proteins were separated by 12.5%

SDS-PAGE (Bio-Rad, Hercules, CA) and transferred onto Hybond-P membrane

(Amersham/GE Healthcare, Piscataway, NJ). The membranes were blocked with

3% milk for 1 hour and incubated with primary antibodies diluted in Blotto. Anti-

26 mouse horseradish peroxidase-conjugated antibody and SuperSignal West Pico

Chemiluminescent substrate (Pierce) were used for signal detection.

2.6 Transfection

For small interfering RNA (siRNA)-mediated knockdown of HuR, LLC-PK1 or

HK-2 cells were plated at 40–50% confluency on six-well plates and transfected transiently with a previously described siRNA [Jeyaraj et al., 2006] of the following sequence: 5’-GGAGGAGUUACGAAGUCUGtt-3’ (sense); 5’-

CAGACUUCGUAACUCCUCCtg-3’ (antisense). HuR siRNA or a non-targeting control (Ambion, Austin, TX) was transfected at 50 nM using Lipofectamine with

Plus reagent (Invitrogen, Carlsbad, CA). RNAi-mediated knockdown of Grb10 was performed by transfecting 100 nM commercially available siRNA (Santa

Cruz) into HK-2 cells using Lipofectamine and Plus reagent (Invitrogen).

For overexpression of a FLAG-tagged HuR cDNA, addition of FLAG to the

COOH-terminal end of the full-length murine HuR coding region was performed using a QuikChange site-directed mutagenesis kit (Stratagene, La Jolla, CA) according to the manufacturer’s recommendations. This cDNA, subcloned in expression vector pcDNA3.1 (Invitrogen), or pcDNA3.1 alone as a control, was stably transfected into LLC-PK1 cells. Transfections were performed using

Lipofectamine and Plus reagent (Invitrogen). Forty-eight hours post-transfection,

cells were treated with 800 μg/ml geneticin (Invitrogen); following expansion of

cells, individual colonies were selected and FLAG-tagged HuR expression was

27 proved by Western blotting and immuno-cytochemical analysis, using an anti-

Flag M2 antibody (Sigma-Aldrich, St. Louis, MO).

2.7 RT-PCR and Immuno-RT-PCR.

An internal standard for competitive RT-PCR of HuR was previously synthesized [Jeyaraj et al., 2006]. An internal standard for Grb10 was synthesized using the same methodology. Total RNA from HK-2 cells was isolated using Trizol (Invitrogen) following the manufacturer’s instructions. A mixture of internal standard RNA and cellular RNA was reverse transcribed using the Super Script cDNA synthesis system (Invitrogen). The resulting cDNA was subjected to PCR using Platinum Taq DNA polymerase (Invitrogen). For HuR,

PCR primers were of the sequences 5’-GGTTATGAAGACCACATGGCCG-3’

(sense) and 5’-AAGCCATAGCCCAAGCTGT-3’ (antisense). Human Grb10 primers were of the sequences 5’-

ACAAGGTGGAGCAGACAAGGTGGAGCAGACACCTC-3’ (sense) and 5’-

GTAAAGAACCCGGCGGTGAGC-3’ (antisense), while murine Grb10 primers were of the sequences 5'-GAATCCAGTGAACTTCTTCCC-3’ (sense) and 5'-

GCGTTGTACTGCTTCTTTCC-3' (antisense).

Immunoprecipitation of HuR followed by RT-PCR of associated mRNAs was performed as previously described [Jeyaraj et al., 2006]. Briefly, HK-2 cells were lysed in CEB buffer (10 mM HEPES, pH 7.5, 3 mM MgCl2, 14 mM KCl, 5%

Glycerol, 0.2% NP-40, 1 mM DTT) containing protease and RNAse inhibitors,

followed by immunoprecipitation of HuR using mouse monoclonal 3A2 (Santa

28 Cruz) and Protein A/G (Thermo Scientific). The resulting precipitate was washed

with CEB buffer, and RNA was extracted using Trizol. Following RNA isolation

and cDNA synthesis, the presence of Grb10 mRNA was assessed using the

PCR primers described above.

2.8 Immunocytochemistry

4 For immunolocalization studies, 4 x 10 LLC-PK1 or HK-2 cells were seeded on glass coverslips in 24-well plates and grown to confluence, at which time the cells were given fresh medium and cultured overnight prior to use. Following the necessary treatments, cells were fixed and permeabilized in 2% formaldehyde in stabilization buffer [Jeyaraj et al., 2006]. Cells were then probed with relevant primary antibodies. Alexa 488-or Alexa 568-conjugated goat anti-mouse secondary antibodies were obtained from Molecular Probes (Eugene, OR). Cells were visualized with a Nikon Eclipse 80i epifluorescent microscope with SPOT software (Diagnostic Instruments, Sterling Heights, MI), or with a Zeiss 510

META laser scanning confocal microscope at the Campus Microscopy and

Imaging Facility at The Ohio State University.

2.9 PCR Arrays

RT2 Profiler PCR arrays and associated reagents were purchased from

SABiosciences/Qiagen. To compare gene expression between control and HuR- suppressed HK-2 cells, total RNA was harvested from cells treated with control oligonucleotide or HuR siRNA 48 hours post-transfection, then subjected to ATP

29 depletion for 6 hours. cDNA synthesis was performed using an RT2 First Strand

kit. PCR arrays were analyzed using a Bio-Rad iCycler iQ Real-time Detection

System in the Plant-Microbe Genomics Facility at The Ohio State University.

Analysis of resulting PCR products was performed using RT2 Profiler PCR Array

Data Analysis software according to SABiosciences/Qiagen protocols.

2.10 Plasmids

Plasmids encoding wild-type and constitutively active Akt were a kind gift of

Dr. Michael Ostrowski (The Ohio State University). The cDNA encoding mouse

Grb10 eta was an I.M.A.G.E. clone obtained from the American Type Culture

Collection (accession number BC016111). Restriction endonuclease XbaI was

used to remove 2434 bases (82.4%) of the Grb10 3’ untranslated region for

experiments testing the role of the 3’ UTR in Grb10 expression. Other plasmids were previously derived in the Lee laboratory.

2.11 In Situ Hybridization

For in situ hybridization of total HuR mRNA, an antisense probe was transcribed in vitro using the MAXIscript system (Ambion). The XhoI-KpnI fragment was subcloned into pBluescript and an antisense probe was made by linearizing the plasmid with XhoI and performing transcription using T7 RNA

polymerase. Alexa 488-labeled UTP (Molecular Probes) replaced unlabeled UTP

in this reaction. The hybridization procedures were essentially as previously

described [Jeyaraj et al., 2006]. The probe was directed to 400 nucleotides

30 present in the coding region. Thus, both HuR mRNA isoforms can be detected

using this probe.

2.12 Sucrose Density Gradients

7 For ribosomal profile protein analysis, 1 x 10 LLC-PK1 cells were seeded in

a 100 mm dish. After 1 or 2 hours of ATP depletion and 4 hours of recovery in

normal growth medium, cells were incubated with 100 μg/ml cycloheximide for 20

minutes. Cells were harvested in PBS, washed twice in PBS and gently lysed on

ice with 500 μl chilled Mg buffer (10 mM HEPES, 10 mM NaCl, 3 mM CaCl2, 7 mM MgCl2, 0.5% NP40) with 1 mM DTT and 100 U/ml RNasin (Promega,

Madison, WI). Cell lysates were centrifuged for 2 minutes at 14,000 × g, then

layered onto a 10 ml linear gradient of 15% to 45% sucrose in 10 mM HEPES, 10

mM NaCl, 3 mM CaCl2, 7 mM MgCl2, 1 mM DTT. Lysates were centrifuged at

36,000 × g for 2.25 hours at 4°C in a Beckman SW41 rotor. Gradients were fractionated and monitored at A254 using an ISCO fractionation system (Lincoln,

NE). Proteins in each fraction were combined as required, precipitated with ice

cold 20% trichloroacetic acid (TCA) for 30 minutes on ice, and centrifuged for 20

minutes at 12,000 RPM at 4°C. Pellets were washed 5 times with acetone, and

then dried for 2 hours in a Speedvac. Pellets were resuspended in SDS loading

dye, boiled for 5 minutes, and loaded on a 12.5% SDS-polyacrylamide gel and

subjected to western blot. RNA was extracted with Trizol and RT-PCR was HuR

mRNA was detected using two different primer sets. Total HuR mRNA was

detected using primer pairs 5’-GGTTATGAAGACCACATGGCCG-3’ (sense) and

31 5’-AAGCCATAGCCCAAGCTGT-3’ (antisense) whereas long form HuR mRNA

was identified by using primer pairs 5’-CGCGCTGAGGAGGAGCC-3’ (sense)

and 5’-CCTGGGTCATGTTCTGAGGGAG-3’UTR (antisense).

2.13 Gel Mobility Shift Assay

Nuclear extracts were prepared from untreated or ATP-

depleted/recovered LLC-PK1 cells as previously described [Lee et al., 1988].

Double-stranded DNA oligonucleotide probes were synthesized that

corresponded to -148 to -109 or -77 to -37 of the porcine HuR 5’-UTR (numbered relative to the translational start). Probes were end-labeled with [γ-32P]-ATP and

T4 polynucleotide kinase using a gel shift assay system (Promega Corp.,

Madison, WI). Fourteen micrograms of nuclear extract were incubated with

labeled probe in the provided binding buffer at room temperature for 20 minutes

prior to separation in a nondenaturing 4% polyacrylamide gel. Supershift assays

were performed by pre-incubating 6 µg of anti-Smad 1/5/8 antibody (Santa Cruz

Biotechnology) with nuclear extracts for 1 hour at 4°C prior to the addition of

radiolabeled probe. Oligonucleotide competition reactions were performed by

pre-incubating the nuclear extracts with unlabeled oligonucleotide for 10 minutes

at room temperature prior to the addition of the labeled probe.

2.14 Chromatin Immunoprecipitation (ChIP)

ChIP assays were performed with the Magna ChIP A kit (Millipore,

Billerica, MA) according to the manufacturer’s protocol. Control and treated LLC-

32 6 PK1 cells (10 x 10 each) were fixed for 10 minutes at room temperature and sonicated according to the manufacturer’s protocol. DNA bound to Smad 1/5/8 was precipitated with anti-Smad 1/5/8 antibody N-18 (Santa Cruz Biotechnology).

To amplify the Smad-binding region in 5’-UTR of the HuR gene, the precipitated

DNA was subjected to PCR using Prime-STAR HS DNA polymerase with GC buffer (Takara Bio Inc.) with the following primers under the following conditions:

5’-GCTGAGGAGGAGCCGC-3’ (sense) and 5’-GGCTGCTCCGGGTCG-3’

(antisense); 3 minutes at 94 °C; and then 30 cycles of 98 °C for 10 seconds, 66

°C for 5 seconds, and 72 °C for 30 seconds; followed by a final extension at 72

°C for 2 minutes. The PCR product was electrophoresed in a 5% polyacrylamide gel and quantified using Image J software. The identities of the resulting bands were confirmed by DNA sequencing.

33

CHAPTER 3

HuR Promotes Cell Survival in ATP-depleted Renal Proximal Tubule Cells

Adapted from Ayupova & Singh et al., Am J Physiol Renal Physiol. 2009 July;

297(1): F95–F105.

3.1 INTRODUCTION

Renal ischemia-reperfusion (IR) injury is a major cause of acute kidney

failure, a clinical condition that despite decades of basic research and technical advances remains associated with high morbidity and mortality [Devarajan,

2006]. Thus understanding the molecular mechanisms of renal cell injury is of

particular importance to the prevention or treatment of acute renal failure.

Various cellular events contribute to IR injury, including reduced cellular

ATP levels, damage to the cytoskeleton, generation of reactive oxygen species,

accumulation of inflammatory cells, and other shifts in cellular activity [Weinberg,

1991]. A number of recent studies have examined whether activation or inhibition

of specific intracellular signaling pathways provides protection from IR injury. It

has been reported that preconditional activation of hypoxia-inducible factors

[Bernhardt et al., 2006], gene silencing of complement 3 and caspase 3 [Zheng

et al., 2006], and adenovirus mediated Bcl-2 and Bcl-XL gene transfer [Chiang et

al., 2005; Chein et al., 2007] ameliorate ischemic acute renal failure, whereas

34 pretreatment with cyclooxygenase (COX)-2 inhibitor augments the degree of

renal dysfunction and injury caused by IR [Patel et al., 2007].

Previous results from the Lee laboratory using the proximal tubule cell line

LLC-PK1 subjected to ATP depletion and recovery suggested a role for human

antigen R (HuR) in protecting kidney epithelia from injury during ischemic stress

[Jeyaraj et al., 2005, 2006]. HuR is a ubiquitously expressed member of the

ELAV family of proteins, participating in posttranscriptional regulation of mRNAs bearing uridine-rich or adenine- and uridine-rich elements (AREs) in their 3’ untranslated sequences [Lopez et al., 2004]. Through identification of its targets,

HuR has emerged as an important regulator in cell division, carcinogenesis, immune responsiveness, and the response to cellular stress [Gallouzi et al.,

2001]. The protective role of HuR during cellular stress is effected, at least partially, through its positive influence on the expression of antiapoptotic genes, as well as its negative effect on proapoptotic genes. HuR was shown to stabilize

SIRT, a major antiapoptotic protein, the human ortholog of Sir2 (the stress- response and chromatin-silencing factor in S. cerevisae) [Abdelmohsen et al.,

2007]. Prothymosin α (ProTα), an inhibitor of apoptosome formation, is regulated by HuR via elevation of its translation [Lal et al., 2005]. More recently, two other

HuR target mRNAs that encode antiapoptotic proteins, Bcl-2 and Mcl-1, have been identified [Abdelmohsen et al., 2007]. Available evidence indicates that

HuR contributes to maintaining their elevated levels, although the precise mode of its action remains to be elucidated. HuR also modulates the expression of numerous proteins that indirectly prevent apoptosis by involvement in regulation

35 of cell signaling [Levy et al., 1998], cell division [Wang et al., 2000], and other functions.

Under normal cellular growth conditions, HuR is localized predominantly in

the nucleus, while stabilization and regulation of ARE-containing mRNA is

associated with translocation of HuR to cytoplasm. In the nucleus, HuR has been

postulated to participate in the processing of pre-mRNA (likely splicing and

export), although these functions remain poorly understood. Recent data about

the function of HuR as an alternative Fas pre-mRNA splicing regulator provide

new insight about HuR as an antiapoptotic and survival factor [Izquierdo, 2008].

However, in spite of the abundance of studies demonstrating an antiapoptotic role for HuR, in some instances a proapoptotic role has been noted [Mazroui et

al., 2008]. It has been suggested that in response to a variety of stresses, HuR

may promote the expression of proapoptotic mRNAs. In one study, HuR cleavage was triggered in response to lethal stress, and the HuR cleavage products were capable of promoting apoptosis [Mazroui et al., 2008]. Therefore, it is important to establish the function of HuR in a given cell type and stress event. In work previously published by the Lee laboratory on the LLC-PK1

proximal tubule cell model, energy depletion at the cellular level caused

detectable net movement of HuR into the cytoplasm where it can stabilize target

mRNAs, followed by net movement of HuR back into the nucleus on reversion to

normal growth medium. Changes observed in HuR protein and mRNA levels

during ATP depletion and recovery also suggested a role for HuR in

preconditioning LLC-PK1 cells to multiple ischemic insults [Jeyaraj et al., 2006]. In

36 this study, the role of HuR expression in the protection of LLC-PK1 cells from

apoptosis was determined to further explore HuR’s function in protecting renal

epithelia from stress.

3.2 RESULTS

3.2.1. Nature and Time Course of Cell Death in ATP-depleted LLC-PK1 Cells.

To determine a role for HuR in renal cell survival, we first needed to establish the nature and time course of death in LLC-PK1 cells undergoing ATP

depletion. This was accomplished by incubating the cells in DMEM base medium

lacking serum and glucose and containing 0.1 μM antimycin, an inhibitor of

mitochondrial ATP synthesis, and measuring cell viability followed by assays for

apoptosis/necrosis. This methodology reduces ATP levels to ~1% normal within

1 hour [Jeyaraj et al., 2006]. Our initial experiments demonstrated that long-term

ATP depletion in our system (>4 hours) induces significant cell death. The

potential role of apoptosis in this death was analyzed using standard indicators

including Hoechst staining, which detects presence of chromatin condensation

and apoptotic bodies, or by measuring activities of caspase 3/7. In some cases,

apoptosis was assayed also by the use of a commercially available fluorescent

TUNEL assay (Roche Applied Science) that permits measurement of cleaved

DNA by fluorescent microscopy. An example of a TUNEL assay is shown in

Figure 3.1. Apoptosis was found to increase from 1 to 6 hours of ATP depletion

but was more pronounced between 4-6 hours. Necrosis was not prevalent until

after 6 hrs of ATP depletion (data not shown).

37

3.2.2. Determination of Role of HuR in Cell Survival

Once we determined the time course of LLC-PK1 cells undergoing apoptosis

during the early hours of ATP depletion, siRNA was used to knock down HuR

expression to determine a potential role of this protein in promoting cell survival.

As previously tested, 1-2 days of siRNA treatment was sufficient to suppress

HuR in LLC-PK1 cells [Jeyaraj et al., 2005]. Following this treatment, cells were

subjected to ATP depletion, and measurement of apoptosis was performed using

caspase 3/7 assays and scoring of apoptotic nuclei.

We have previously demonstrated the ability to use siRNAs to suppress

HuR in LLC-PK1 cells under normal growth conditions [Jeyaraj et al., 2006], and

Figure 3.2A demonstrates that knockdown was similarly efficient in cells

undergoing ATP depletion (knockdown averaged about 75%). Measurement of

caspase 3/7 activation (Figure 3.2B, left) or quantitation of condensed/fragmented nuclei (Figure 3.2B, right) demonstrated that ATP depletion of control cells resulted in increasing levels of apoptosis over a 6 hour

period. Suppression of HuR expression by siRNAs resulted in even greater apoptosis during ATP depletion. Similar results were obtained using the human proximal tubule cell line HK-2 (data not shown). These results indicate the protective role of HuR in proximal tubule cells during ATP depletion.

3.2.3. Overexpression of HuR Promotes Cell Survival

38 For overexpression of HuR in LLC-PK1 cells, a murine HuR cDNA was epitope tagged so it was distinguishable from the endogenous (porcine) LLC-PK1

HuR by Western analysis. For this purpose, a FLAG tag (8 amino acid residues) was appended to the C-terminus of HuR. The FLAG epitope tag was chosen because a previously made HuR-FLAG hybrid was shown to traffic appropriately in NIH 3T3 cells [Lal et al., 2005]. The same epitope tag was inserted at the C- terminal end of the coding region while still maintaining the integrity of the 3’

UTR, using the Quick-Change II site directed mutagenesis system (Stratagene).

Clonal LLC-PK1 lines expressing the HuR-FLAG hybrid or empty pcDNA3.1

vector were then made by isolating stably transfected cells expressing a

geneticin resistance marker. Following expansion of cells, individual colonies

were selected and FLAG-tagged HuR expression was proved by Western

blotting and immunocytochemical analysis. With the use of commercially

available anti-FLAG antibody, products of exogenously expressed clones were

detected. Expression and correct localization of FLAG-tagged HuR was checked

by immunocytochemistry. As shown in Figure 3.3, LLC-PK1 cells stably

expressing empty vector or FLAG-tagged HuR were grown under both normal

and ATP-depleted conditions. The vector-only cells demonstrate the distribution

of HuR seen in untreated LLC-PK1 cells; namely, that in normal growth medium,

HuR is primarily nuclear, but upon ATP depletion, its distribution becomes more

cytoplasmic. Similarly, detection of the FLAG tag in the HuR-overexpressing lines

shows that the tagged version also is distributed primarily in the nucleus in

normal growth medium but becomes cytoplasmic under ATP depletion

39 conditions. These results demonstrate appropriate trafficking of the HuR-FLAG hybrid protein.

FLAG-tagged HuR overexpressing clones were used to study the effect

of heightened levels of HuR on apoptosis. Although only one clonal line is

discussed here, a second line produced very similar results (data not shown).

As shown in Figure 3.4A, expression of HuR in these lines was approximately

2-3-fold times normal. These cells were subjected to ATP depletion for

different times and relative caspase 3/7 activity was measured (Figure 3.4B,

left), as well as numbers of apoptotic nuclei (Figure 3.4B, right). A decrease in

apoptosis was noted in HuR overexpressing cell lines in comparison to a line

stably transfected with empty vector. This further establishes the protective

role of HuR under conditions of ATP depletion.

3.2.4. HuR Promotes Cell Survival by Inhibiting Both Intrinsic and Extrinsic

Apoptosis Pathways.

Understanding the molecular events that lead to apoptosis in response to

ATP depletion, and how HuR delays apoptotic death, provides an understanding

of HuR’s function in renal ischemia related apoptosis. In order to observe

whether HuR mediates its anti-apoptotic effects through extrinsic and/or intrinsic

apoptotic pathways during ATP depletion, HuR was suppressed in LLC-PK1 cells

using siRNA HuR. Following the suppression, cells were ATP depleted for

various time points, and relative caspase 9 (intrinsic pathway, Figure 3.5A) and

caspase 8 (extrinsic pathway, Figure 3.5B) activities were measured.

40 Suppression of HuR expression by siRNAs resulted in greater apoptosis of both

pathways during ATP depletion. These results indicate that both pathways are

activated during ATP depletion of renal proximal tubule cells, and that HuR play a

protective role in each of them.

3.2.5. HuR Stabilizes Cell Survival mRNAs Bcl-2 and Hsp70 under Cell

Stress.

We wished to determine whether HuR might play a role in modulating

expression levels of proteins known to play protective roles in in vivo and in vitro

models of renal ischemia. The expression of Bcl-2, which is upregulated in

regenerating proximal tubule cells and is protective against renal ischemic injury

[Basile et al., 1997; Suzuki et al., 2008], was previously shown to be modulated

by HuR [Abdelmehsen, 2007]. As demonstrated in Figure 3.6, siRNA-mediated

suppression of HuR resulted in diminished Bcl-2 expression during ATP depletion in LLC-PK1 cells. In addition, suppression of HuR mildly diminished expression of Hsp70, a heat shock protein known to be upregulated primarily in

proximal tubule cells by renal ischemia and involved in a cellular-protective

response [Smoyer, 2000]. This is consistent with other studies demonstrating the

Hsp70 mRNA to be a target of HuR binding [Lopez et al., 2007]. Thus, HuR

appears to play a protective role in energy-depleted renal epithelia in vitro.

3.3. CONCLUSION

41 IR-induced changes include damage of renal tubular epithelial cells and especially cells of proximal tubules, known to be more susceptible to injury in this setting. Using an ATP depletion model in the proximal tubule cell line LLC-PK1, we have suggested a role for HuR in protecting kidney epithelia from injury during ischemic stress. Based on pull-down of HuR-mRNA complexes followed by microarray analysis, HuR has been predicted to bind several thousand cellular mRNAs [Lopez et al., 2004]. Here, we have demonstrated that suppression of

HuR in LLC-PK1 cells diminishes expression of at least two molecules key to cell survival following renal ischemia and reperfusion, Bcl-2 and Hsp70. With maturation of the kidney, Bcl-2 expression is detected in the parietal epithelium of

Bowman’s capsule, in the distal convoluted tubule and loop of Henle, and in widely scattered cells within proximal convoluted tubules and papillary collecting ducts. Bcl-2 resides in the mitochondria and prevents activation of the effector caspases. In addition, it is known that over-expression of Bcl-2 can block both apoptosis and necrosis [Supavaken, 2003], and protect ischemic tissue against reperfusion induced stress [Gobe, 1999].

Hsp70 is a stress response protein that interacts with cytoskeletal elements during the restoration of proximal tubule cell structure and polarity after renal ischemia [Bettina et al., 2000]. Hsp70 has been demonstrated to be protective in renal tubular cell apoptosis that is induced by inflammatory cytokine or ATP depletion [Supavaken, 2003]. In our findings, the level of both Hsp70 and Bcl-2 anti-apoptotic proteins decreased upon suppression of HuR (Figure 3.6). Taken together, our results show that HuR plays an antiapoptotic role in a proximal

42 tubule cell line, which suggests protective and preconditioning roles of HuR in renal ischemic injury.

43

Figure 3.1. Time Course of Apoptosis in LLC-PK1 cells during ATP Depletion. TUNEL assays were used to determine levels of apoptosis in LLC-

PK1 cells undergoing 30 minutes to 6 hours of ATP depletion. Apoptosis, as indicated by the fluorescent green labeling, steadily increased during this time.

44

Figure 3.2. Effects of HuR Knockdown on Apoptosis in ATP-depleted LLC-

PK1 cells. (A) Knockdown of HuR in LLC-PK1 cells with a small interfering RNA against HuR prior to ATP depletion resulted in an average knockdown of ~75%, as indicated by Western blot. Levels of β -actin are presented as a control for loading. The increase in HuR protein seen during ATP depletion was previously described [Jeyaraj et al., 2006]. (B) Knockdown of HuR resulted in increased apoptosis of LLC-PK1 cells during ATP depletion, as assayed by caspase 3/7 activity (left) or numbers of apoptotic nuclei (right). Values are mean + standard deviation; n = 3. *P < 0.05; **P < 0.01.

45

Figure 3.3. Distribution of Native and FLAG-tagged HuR in Stably

Transfected LLC-PK1 Cells. LLC-PK1 cells stably expressing a FLAG-tagged HuR construct were immunolabeled with an anti-FLAG antibody (top row). When cells were cultured in normal growth medium, the FLAG-HuR was primarily nuclear in distribution. However, ATP depletion for 4 hours resulted in movement of FLAG-HuR into the cytoplasm. The distribution of FLAG-HuR in these cells is consistent with that of endogenous HuR in LLC-PK1 cells transfected with an empty vector (bottom row) or untransfected cells [Lal et al., 2005].

46

Figure 3.4. Effects of HuR Overexpression on Apoptosis in ATP-depleted

LLC-PK1 cells. (A) Stable transfection of FLAG-tagged HuR into LLC-PK1 clonal lines resulted in increased overall HuR levels, as indicated by Western blot. In two separate lines, HuR expression was increased 2-3 fold. The lower panel indicates β -actin levels as a loading control. (B) HuR overexpression resulted in decreased apoptosis of LLC-PK1 cells during ATP depletion, as assayed by caspase 3/7 activity (left) or numbers of apoptotic nuclei (right). Values are mean + standard deviation; n = 3. *P < 0.05; **P < 0.01.

47

Figure 3.5. Effects of HuR Knockdown on Intrinsic and Extrinsic Pathways of Apoptosis. LLC-PK1 cells were treated with an siRNA to HuR or control oligonucleotides prior to ATP depletion for up to 6 hours. Relative caspase 8 or 9 activity was measured using a Caspase Glo Assay Kit (Promega). Knockdown of HuR resulted in increased apoptosis through both the intrinsic (caspase 9) and extrinsic (caspase 8) pathways. Values are mean + standard deviation; n = 3. *P < 0.05.

48

Figure 3.6. Knockdown of HuR in ATP-depleted LLC-PK1 Cells Results in Suppression of Cell Survival Proteins. HuR expression was knocked down in LLC-

PK1 cells with siRNAs prior to ATP depletion for up to 6 hours. Bcl-2 and Hsp70 levels were assessed by Western analysis. Knockdown of HuR resulted in loss of expression of both survival proteins. β-actin levels were assessed as loading controls.

49

CHAPTER 4

HuR mediates anti-apoptotic effect by amplifying Akt signaling through a positive feedback loop, via Grb10

4.1 INTRODUCTION

HuR (human antigen R) is a ubiquitously expressed regulator of post- transcriptional processing, with effects on pre-mRNA splicing, mRNA stability, and translation. HuR has been estimated to regulate ~8% of the human transcriptome, with broad effects on cell cycle, apoptosis, and inflammatory responses [Khabar, 2005]. Its most profound effects are on mRNA stability, in which it binds to adenine- and uridine-rich elements (AREs) in the 3’ untranslated regions of mature mRNAs and slows their rates of degradation. Regulation of

HuR expression must be very tightly controlled, as loss of HuR expression results in rapid cell death [Ghosh et al., 2009; Katsanou et al., 2009], while overexpression by only a few-fold results in tumorigenicity [Danilin et al., 2010;

Denkert et al., 2004a; Denkert et al., 2004b; Lopez de Silanes et al., 2003; Lopez de Silanes et al., 2005; Mazan-Mamczarz et al., 2008; Nabors et al., 2001]. We have demonstrated that in cultured renal proximal tubule cells, HuR stabilizes mRNAs and prevents apoptosis resulting from ATP depletion [Ayupova and

Singh et al., 2009; Jeyaraj et al., 2005]. In rat kidneys, ischemia-reperfusion injury results in overall increases in HuR mRNA throughout the nephron, but HuR

50 protein levels are increased only in proximal tubules [Ayupova and Singh et al.,

2009]. This distinction is notable, as proximal tubule cells are highly sensitive to

ischemic injury.

HuR’s capacity to bind mRNAs is enhanced during cell stress, when it switches from a typical nuclear steady-state location to the cytoplasm [Atasoy et

al., 1998; Fan and Steitz, 1998]. Stresses that trigger HuR activation include

energy depletion, ultraviolet light, hypoxia, nutrient deprivation, and heat shock,

among others. HuR has been shown to promote the mRNA stability and/or

translation of several anti-apoptotic proteins including Bcl-2, Mcl-1, prothymosin-

α, and XIAP. In addition, it was demonstrated that HuR promotes alternative

splicing of the mRNA for the apoptosis receptor Fas such that the resulting

protein no longer retains its pro-death functions [Izquierdo, 2008].

The PI3K/Akt pathway has been demonstrated to be critical to cell survival

and repair in multiple tissue injury models, including the kidney. Renal ischemia/reperfusion (IR) was shown to transiently increase Akt activation

[Andreucci et al., 2003], which was further demonstrated to play a protective role in IR-injured kidney function [Satake et al., 2008]. The PI3K/Akt pathway also has been shown to protect against acute kidney injury induced by cisplatin toxicity [Kuwana et al., 2008]. Akt, a family of highly related serine/threonine kinases, protects against apoptosis through phosphorylation of multiple proteins involved in regulating cell survival, including Bcl-2 family members, caspases and caspase inhibitors, the mTOR signaling pathway, and FoxO transcription factors.

Previous studies have suggested interaction between Akt signaling and HuR

51 function. In gastric tumor cells, it was shown that Akt activation could promote

HuR expression through stimulation of an NF-κB element in the HuR promoter

[Kang et al., 2008]. Conversely, it was suggested that HuR was required for Akt activation in renal tubule cells [Danilin et al., 2010]. Therefore, the precise interaction of HuR with the PI3K/Akt pathway is unclear.

Grb10 is a member of a family of adaptor proteins (including Grb7 and

Grb14) that functions downstream of multiple receptor tyrosine kinases such as

the insulin and insulin-like growth factor-I receptor [Liu and Roth, 1995; Morrione

et al., 1996], growth hormone receptor [Moutoussamy et al., 1998], and the Ret

[Pandey et al., 1995], among others. Grb10 may also

interact with a number of non-receptor kinases, including Akt. It has been

proposed that Grb10 stimulates Akt function by translocating Akt to the plasma

membrane where it is phosphorylated and activated by PI3 kinase [Jahn et al.,

2002]. Here we show that HuR binds Grb10 mRNA and promotes Grb10

expression, leading to their participation in a positive feedback loop that amplifies

the pro-survival functions of Akt signaling in renal proximal tubule cells.

4.2. RESULTS

4.2.1. HuR Expression is Regulated by PI3K/Akt Activity in Proximal Tubule

Cells.

Numerous previous reports have established the importance of PI3K/Akt

activity in cell survival in energy-depleted renal epithelia [Andreucci et al., 2003;

Chen et al., 2008; Joo et al., 2006; Loverre et al., 2004; Sharples et al., 2004; Xie

52 et al., 2006]. To confirm these findings in vitro, the human proximal tubule cell line HK-2 was subjected to ATP depletion in the presence or absence of PI3K inhibitor LY-294002, and apoptosis was measured through assessment of caspase 3/7 activity. Figure 4.1A demonstrates that over the course of 6 hours, activity of these caspases increased approximately 7-fold and that the magnitude of this response was almost doubled by inhibition of PI3K signaling. In addition,

Figure 4.1B demonstrates that ATP depletion resulted in changes in the distribution of Akt. Control HK-2 cells demonstrated a primarily nuclear distribution of Akt, as has been demonstrated for a number of cell types including

HEK293, HeLa, PC12, and cardiomyocytes [Martelli et al., 2006; Miyamoto et al.,

2009; Wang and Brattain, 2006]. In contrast, ATP-depleted cells demonstrate a clear accumulation of Akt at the cell periphery (arrows), indicative of increased activity. ATP depletion also induces an increased in pAkt levels, as assessed by

Western blots (below). These results confirm a role for PI3K/Akt signaling in cell survival in HK-2 cells undergoing energy depletion.

PI3K/Akt signaling was previously shown to increase HuR expression in gastric tumor cells [Kang et al., 2008]. Because HuR, like PI3K/Akt, promotes cell survival in energy-depleted renal epithelia [Ayupova and Singh et al., 2009], we performed experiments to determine whether PI3K/Akt signaling enhanced

HuR expression in proximal tubule cells. The HK-2 cell line was ATP depleted in the presence or absence of LY-294002 and HuR expression levels were determined. As was previously demonstrated in the LLC-PK1 cell line [Jeyaraj et al., 2006], ATP depletion of HK-2 cells results in increased HuR protein

53 expression without increased HuR mRNA levels. Here, competitive RT-PCR

revealed that HuR mRNA levels did not change over 6 hours of ATP depletion in

control cells, but inhibition of PI3K by LY-294002 resulted in loss of HuR mRNA

expression (Figure 4.1C, top row). Figure 4.1C, middle row, demonstrates that

HuR protein levels increased during ATP depletion in HK-2 control cells.

However, consistent with the RT-PCR results, LY-294002 treatment caused a

dramatic loss of HuR protein. These results demonstrate that PI3K activity

regulates HuR expression in proximal tubule cells under both normal (timepoint

0) and energy-depleted conditions. Finally, to determine whether PI3K regulates

HuR expression via Akt, HK-2 cells were transfected either with wild-type or

constitutively active (myristoylated) Akt1. As shown in 4.1D, total Akt levels were

increased when either wild-type or CA-Akt was overexpressed. However, only

the constitutively active form caused an increase in HuR protein levels, indicating that PI3K/Akt signaling plays an important role in regulating HuR expression in proximal tubule cells under normal and stressed conditions.

4.2.2. NF-κB is Critical for HuR Expression during Normal Growth.

To determine whether PI3K/Akt signaling regulates HuR expression through

the NF-κB signaling pathway, normally growing HK-2 cells were treated with

inhibitor NF-κB inhibitor BAY11-7082, and HuR levels were assessed. Both

Western blot (Figure 4.2A) and competitive RT-PCR (Figure 4.2B) demonstrate

that inhibition of NF-κB activity in normally growing cells results in dramatic

decreases in HuR expression at both the mRNA and protein levels. To

54 demonstrate the relationship between Akt and NF-κB relative to HuR expression,

HK-2 cells overexpressing either WT or CA Akt were treated with NF-κB inhibitor

BAY11-7082. As shown in Figure 4.2C, inhibition of NF-κB resulted in loss of

HuR, indicating that Akt is upstream of NF-κB in this signaling pathway.

We were unable to assess the effects of NF-κB inhibition in cells undergoing

ATP depletion, as the two treatments together resulted in rapid (within minutes)

cell death. However, as we previously published, HuR protein levels increase during ATP depletion without an associated change in HuR mRNA levels [Jeyaraj et al., 2006]. Therefore, it seems unlikely that NF-κB signaling plays a significant

role in modulating HuR levels during ATP depletion.

4.2.3. Akt Activity but not Akt Expression is Regulated by HuR Levels in

Proximal Tubule Cells.

The previous data demonstrate that HuR levels are increased by PI3K/Akt

signaling in proximal tubule cells. Interestingly, other studies have suggested

that this relationship may be reciprocal; that is, that HuR can also modulate Akt

activity [Danilin et al., 2010; Meng et al., 2002]. Indeed, our own preliminary

studies using PCR arrays to examine changes in gene expression with HuR

knockdown suggested that HuR levels do affect Akt signaling (not shown).

Therefore, we performed Western analysis in our cell model to determine

whether siRNA-mediated suppression of HuR affected Akt expression or activity.

Figure 4.3A, top row, illustrates that HuR protein expression, which increases

during ATP depletion, can be strongly inhibited by HuR siRNAs in HK-2 cells.

55 Figure 4.3A also demonstrates that Akt is activated during ATP depletion and

that HuR knockdown almost completely abolishes this activation. In contrast,

total Akt expression is unaffected by the knockdown, demonstrating that HuR

modulates activation, rather than the expression, of Akt.

4.2.4 Expression of Adaptor Protein Grb10 is Regulated by HuR Levels in

Proximal Tubule Cells.

Because HuR is a post-transcriptional regulator of gene expression and not

a kinase, it follows that HuR’s effects on Akt activity are likely to be produced by alteration in expression of an upstream activator of Akt. To determine what members of the PI3K/Akt signaling pathway might be regulated by HuR, PCR array analysis was performed on control HK-2 cells and those in which HuR expression was suppressed by RNAi. In two independent experiments, the mediator of Akt signaling most strongly affected by HuR suppression showed at least a 10-fold loss of expression in HuR-suppressed cells. This protein, Grb10, is a member of the Grb7 family of adaptor proteins that has been shown to be a key regulator of PI3 kinase activity downstream of the insulin and insulin-like growth factor-1 receptor tyrosine kinases [Lim et al., 2004; Riedel, 2004]. To confirm the PCR array analysis, Grb10 mRNA levels were compared in control cells and cells in which HuR was suppressed by RNAi. As shown in Figure 4.3B,

Grb10 mRNA levels increased throughout ATP depletion; however, suppression of HuR inhibited Grb10 expression.

56 This result suggests that Grb10 mRNA may be a target of HuR-mediated

post-transcriptional control. To determine whether HuR binds Grb10 mRNA,

HuR from control or ATP-depleted cells was immunoprecipitated, the co-

precipitating mRNAs were isolated, and the presence of Grb10 mRNA was

assessed by RT-PCR. As shown in Figure 4.3C, under both normal and

stressed conditions, HuR was capable of co-precipitating Grb10 mRNA. An

increase in HuR-associated Grb10 was noted in stressed cells, as is consistent

with HuR’s protective role (in 3 independent experiments, an average increase of

66 + 21% over control cells). A search of the ARED databases [Bakheet et al.,

2006; Halees et al., 2008], which identify ARE-containing mRNAs through in

silico analysis, indicated that Grb10 contains at least one of these sequences.

To determine whether the 3’ UTR does indeed contain a functional ARE, full- length mouse Grb10 or a mutant lacking most of the 3’ UTR was transfected into

HK-2 cells in the presence or absence of exogenous HuR. As shown in Figure

4.3D, the exogenous mouse Grb10 mRNA and protein were then detected by

RT-PCR (top row) or in Western blots using an antibody specific to the mouse protein (middle row). These experiments show that intense bands were detected in cells overexpressing wild-type Grb10 (lanes 3 and 5) but not truncated Grb10

(lanes 4 and 6). This result is consistent with a positive role for the Grb10 3’ UTR in its expression. Additionally, Grb10 levels were highest when co-transfected with HuR (lane 5). These results, coupled with the previous figure showing

HuR’s ability to bind Grb10 mRNA, demonstrate that HuR plays a positive role in

promoting Grb10 expression.

57

4.2.5. Grb10 Stimulates Akt Activation and HuR levels.

As described above, Grb10 has been implicated in some cell types as a positive regulator of cell survival [Lim et al., 2004; Riedel, 2004]. To determine whether Grb10 has any effects on cell survival and the PI3K/Akt signaling pathway in proximal tubule cells, RNAi was used to knock down its expression.

Four alternately spliced Grb10 isoforms with yet unknown specific functions are present in human cells. The siRNAs used in this study were designed to knock down all isoforms. Figure 4.4A demonstrates that Grb10 mRNA can be strongly suppressed in HK-2 cells using nanomolar levels of siRNA. This loss of Grb10 mRNA results in a corresponding loss of Grb10 protein (Figure 4.4B). To determine whether Grb10 has effects on cell survival, HK-2 cells treated with either Grb10 siRNA or a control oligonucleotide underwent ATP depletion, and apoptosis was measured by assays for caspase 3/7 activation. As shown in

Figure 4.4C, ATP depletion in control cells resulted in a ~10-fold increase in caspase 3/7 activity over 6 hours. However, suppression of Grb10 expression increased this response over time, and after 6 hours, caspase 3/7 activity was

~50% higher in the siRNA-treated cells than the control cells. Thus, Grb10 plays an important role in cell survival during energy depletion.

Grb10 was previously shown to be a positive regulator of Akt signaling downstream of PI3 kinase activity [Jahn et al., 2002]. To test whether Grb10 has a similar effect in proximal tubule cells, Grb10 was knocked down by RNAi in cells undergoing ATP depletion, and total and activated Akt levels were assessed

58 by Western blot. As shown in the top row of Figure 4.5A, ATP depletion of

control cells induced Akt phosphorylation, as previously demonstrated in Figure

4.3A. The same row shows that suppression of Grb10 resulted in a dramatic decrease in Akt activation in non-stressed cells and prevented any increase in activation during ATP depletion. This was not due to alterations in total Akt levels, however, as these were unchanged by loss of Grb10 (Figure 4.5A, middle row). Interestingly, siRNA-mediated knockdown of Grb10 also resulted in severe loss of HuR (Figure 4.5A, bottom row). This result suggests that HuR participates in Akt signaling in a positive feedback loop, promoting expression of

Grb10, which then amplifies Akt activation through NF-κB to further enhance

HuR expression. To determine whether knockdown of Grb10 diminishes HuR

expression at the level of mRNA, control cells or those treated with Grb10 siRNA were assessed for HuR mRNA levels by competitive RT-PCR. As shown in

Figure 4.5B, suppression of Grb10 did indeed result in the loss of HuR mRNA.

This result is consistent with Grb10 activating Akt signaling, which then

stimulates expression of HuR.

Finally, we examined the relative distributions of Grb10 and phosphorylated

Akt. Figure 4.5C demonstrates an almost complete overlap of pAkt and Grb10 in

the nucleus under normal growth conditions. In addition, ATP depletion results in

Grb10, along with pAkt, being re-distributed to the cell periphery. As expected

from the results in Figure 4.5A, knockdown of Grb10 resulted in an almost

complete loss of pAkt as assayed by immunocytochemistry (not shown). These

59 results are consistent with a role for Grb10 in assisting in translocation of pAkt to

the plasma membrane in renal epithelia.

4.3 CONCLUSION

The studies described here suggest a central role for HuR within a positive

feedback loop that amplifies Akt signaling in proximal tubule cells. A schematic

of this proposed feedback loop, which is based both on the data presented here

as well as work published in the literature, is shown in Figure 4.6. In brief, we

propose that HuR expression stabilizes Grb10 mRNA, resulting in cytoplasmic

relocalization of pAkt, where it can increasingly stimulate NF-κB activity. NF-κB

activity, then, promotes HuR transcription. We hypothesize that this feedback

loop plays a key role in maintaining pAkt activity under normal growth conditions.

However, we were unable to determine whether NF-κB signaling is important for

HuR levels during energy depletion, since inhibiting this pathway during ATP

depletion rapidly killed the cells. However, this seems unlikely, as we found that

during ATP depletion, HuR protein levels increased without an accompanying

increase in HuR mRNA (this study and [Jeyaraj et al., 2006]). It is possible that

during stress, NF-κB activity is required to just maintain HuR mRNA levels, but it

is clear that an additional level of translational control is responsible for increases

in HuR protein. However, we demonstrated that PI3K/Akt signaling is indeed

involved in increased HuR protein expression during stress, as the PI3K inhibitor

LY-294002 strongly diminished HuR protein levels. Further complicating this

regulation is our finding that HuR mRNA is expressed as two isoforms with

60 different 5’ untranslated regions and different translatabilities in rabbit reticulocyte lysates [Ayupova et al., 2009; Jeyaraj et al., 2010]. Our preliminary studies indicate that these are differentially translated during normal growth and cell

stress (not shown), so experiments are underway to define the timing of these events.

These studies demonstrate how HuR plays a central, positive role in

proximal tubule cell survival through the activities of Akt.

61

Figure 4.1. HuR Levels are Regulated by PI3K/Akt Signaling. (A) Apoptosis levels were assessed by measurements of caspase 3/7 activity in HK-2 cells treated either with vehicle or PI3K inhibitor LY-294002. *, P< 0.05; **, P<0.005. (B) Control or ATP-depleted HK-2 cells were immunolabeled with an antibody against Akt. Arrows indicate plasma membrane distribution of Akt. (C) The effects of PI3K inhibition on HuR levels were measured by competitive RT-PCR (top panel) or Western blot (2nd panel). β -actin was detected as a loading control. For competitive RT-PCR, relative HuR levels are indicated by comparing ratios of the top HuR band to the bottom internal standard band. (D) HK-2 cells were either left untransfected (unt), transfected with an empty expression vector (mock) or transfected with wild-type (WT) or constitutively active (CA) Akt1. Western analysis shows levels of total Akt, HuR, or β -actin as a loading control.

62

Figure 4.2. HuR Levels are Regulated by NF-κB Signaling. The effects of NF- κB inhibition on HuR expression were demonstrated by treatment of HK-2 cells with BAY11-7082. HuR levels were assessed by Western blot (A) and competitive RT-PCR (B). (C) HK-2 cells overexpressing wild-type (WT) or constitutively active (CA) Akt1 were treated with vehicle or BAY11-7082. HuR levels were assessed by Western analysis.

63

Figure 4.3. HuR Regulates Akt Activation and Grb10 Expression. (A) Western analysis demonstrates that ATP depletion increases HuR protein levels and Akt phosphorylation. siRNA-mediated knockdown of HuR strongly inhibits Akt activation without affecting total Akt levels. β-actin is shown as a loading control. (B) Competitive RT-PCR demonstrates that Grb10 mRNA levels increase during ATP depletion, but are diminished by HuR knockdown. (C) HuR was immunoprecipitated from ATP-depleted or control cells, and associated RNA was purified. The presence of Grb10 mRNA was assessed by RT-PCR. Controls demonstrate specificity by showing that a lack of HuR antibody or reverse transcriptase (RT) in the procedure fails to produce Grb10 mRNA. (D) The role of the Grb10 3’ UTR in controlling its expression was demonstrated by both RT- PCR (top row) and Western blot (middle row). Analysis of β-actin levels is included as a loading control for the Western blot (bottom row). Lane 1: untransfected cells; lane 2: cells transfected with empty expression vector; lane 3: transfection with full-length murine Grb10; lane 4: transfection with murine Grb10 lacking 3’ UTR sequences; lane 5: transfection with full-length murine Grb10 plus HuR; lane 6: transfection with murine Grb10 lacking 3’ UTR sequences plus HuR.

64

Figure 4.4. siRNA-mediated Knockdown of Grb10 Increases Apoptosis in ATP-depleted HK-2 Cells. (A) Competitive RT-PCR demonstrates efficient knockdown of Grb10 mRNA with nanomolar levels of siRNA. (B) Western analysis demonstrates efficient knockdown of Grb10 protein during ATP depletion. β-actin levels are assessed as a loading control. (C) Caspase 3/7 assays show that knockdown of Grb10 increases apoptosis during ATP depletion. *, P < 0.05; **, P< 0.01.

65

Figure 4.5. Knockdown of Grb10 inhibits Akt activation and HuR expression. (A) Western analysis of HK-2 cells treated with control oligonucleotides or Grb10 siRNAs show that suppression of Grb10 results in strongly diminished Akt activation and HuR protein expression. (B) Competitive RT-PCR demonstrates that knockdown of Grb10 decreases HuR expression at the mRNA level. (C) Immunocytochemical analysis demonstrates almost complete overlap between Grb10 and pAkt distribution in both control and ATP- depleted cells. ATP depletion results in movement of both proteins to the plasma membrane.

66

Figure 4.6. Schematic of HuR’s role in promoting a positive feedback loop of Akt signaling. Based on data presented here and in the literature, we propose the following positive feedback loop for HuR in Akt signaling. Green arrows represent the pathway found in normally growing cells, while red arrows represent the pathway found in ATP-depleted cells. In normally growing cells, (1) Akt activation stimulates NF-κB activity; (2) NF-κB promotes HuR transcription; (3) HuR binds to and stabilizes Grb10 mRNA, resulting in increased Grb10 expression; (4) Grb10 activates Akt by aiding in its transport to the plasma membrane where it is activated by PI3K. In ATP-depleted cells, the relationship between pAkt activation and HuR levels are not clear, but does not appear to involve NF-κB signaling.

67

CHAPTER 5

Transcriptional and Translational Control of HuR

Some portions adapted from Jeyaraj et al., J. Biol. Chem. 285(7): 4432, 2010

5.1 INTRODUCTION

Cellular levels of HuR must be tightly regulated. We and others showed that

decreased levels of HuR result in increased apoptosis, while levels only a few-

fold over normal can result in tumor formation. However, at the same time, HuR expression must be dynamic to counteract effects of various cellular stresses. An interest of the Lee laboratory has been the regulatory mechanisms that control

HuR expression [Jeyaraj et al., 2006]. We found that ATP depletion and recovery of LLC-PK1 cells results in an unusual decoupling of HuR protein and

mRNA levels. First, ATP depletion induces a slow translation-mediated increase

in HuR protein, without an accompanying increase in HuR mRNA. When cells

are allowed to recover from ATP depletion, HuR protein levels revert to normal,

but HuR transcription is mildly induced. A second insult after recovery results in

a rapid increase in HuR protein without increased mRNA production.

Significantly, this increase in protein is dependent on transcriptional activity

during the prior recovery period. These findings are reminiscent of ischemic

preconditioning; the phenomenon whereby brief ischemic episodes render cells

68 resistant to subsequent ischemic injury [Jeyaraj et al., 2006]. Therefore, the mechanisms by which HuR transcription and translation are regulated may have great impact on protection and recovery of cells from various stresses. The experiments described in this chapter elucidate many of the transcriptional and translational regulatory processes that affect levels of HuR expression during normal growth and stress conditions.

5.2 RESULTS

5.2.1 Gel Shift Assay and ChIP Confirms Binding of Smad 1/5/8 to HuR

Promoter Elements

Results from the Lee laboratory demonstrated that under basal conditions,

HuR mRNA is expressed in two forms: one that contains an approximately 20- base 5'-untranslated region (UTR) sequence with a very low G+C content and one that contains an approximately 150-base, G+C-rich 5'-UTR that is inhibitory to translation in rabbit reticulocyte lysates. Recovery from cellular stresses induced increased expression of the shorter, more translatable transcript and decreased expression of the longer form [Ayupova and Singh et al., 2009]. We predict that expression of the shorter mRNA is induced during recovery from stress to allow rapid translation of HuR upon potential subsequent insults.

Therefore, identification of the promoter elements that drive transcription of this mRNA may give clues to the regulatory mechanisms that protect cells from repetitive insults. Promoter analysis of HuR upstream regions revealed sequences necessary for regulation of the shorter mRNA. Through use of

69 promoter-luciferase reporter constructs, it was determined that sequences critical for transcription of the short mRNA resided within the exon encoding the 5’ UTR of the long mRNA. Within the G+C-rich 5'-UTR exist multiple overlapping copies of the alternate Smad 1/5/8-binding motif GCCGnCGC.

To determine whether the putative Smad 1/5/8-binding sites in the HuR 5’-

UTR are indeed capable of binding Smads, gel mobility shift assays were performed on nuclear extracts derived from both untreated LLC-PK1 cells and those that were ATP-depleted and allowed to recover. Figure 5.1A shows an example of this method, using a porcine probe corresponding to the sequence -

148 to -109 (relative to the start of translation) that encompasses five perfect and two imperfect copies of the GCCGnCGC motif. As shown, one major band was shifted in the presence of nuclear extracts. This band was routinely more intense in extracts from ATP depleted/recovered cells than untreated cells (an increase of 87 + 18%; n=5). The addition of an excess of unlabeled probe caused loss of this band, whereas addition of an irrelevant unlabeled oligonucleotide did not.

Preincubation of extracts with antibody against Smads 1/5/8 resulted in the disappearance of the band, rather than a supershift, indicating potential interference of the antibody with the probe-binding site (data not shown).

Therefore, we performed chromatin immunoprecipitation (ChIP) assays to confirm the identities of the relevant bands. As shown in the bottom panel of

Figure 5.1B, immunoprecipitation of chromatin with an anti-Smad 1/5/8 antibody isolated a fragment containing the HuR 5’-UTR sequences. Consistent with the gel mobility shifts, Smads 1/5/8 roughly doubled their association with the HuR

70 5’-UTR following ATP depletion/recovery (an increase of 86 + 15%; n = 3). These

results demonstrate that the HuR 5’-UTR is indeed capable of binding R-Smads

and that binding activity is increased in cells subjected to ATP depletion and

recovery.

5.2.2 Confirmation of the Presence of BMP-7 Receptors in LLC-PK1 cells

It is of interest that the stress and recovery from ATP depletion induces

transcription of a readily translatable HuR mRNA driven by the family of Smad

1/5/8 transcription factors. Activation of Smads 1/5/8 occurs through signaling by

bone morphogenetic proteins (BMPs). One BMP isoform in particular, BMP-7,

plays a key role in kidney development, as BMP-7-null mice exhibit a lack of

renal mesenchymal differentiation and an absence of nephrons [Dudley et al.;

Luo et al., 1995]. Further, recent reports have demonstrated that BMP-7 plays

an important role in recovery from multiple types of kidney injury, including

damage from ischemia and reperfusion [Zeisberg et al., 2003; Hruska et al.,

2000].

In order to determine whether BMP receptors are present in LLC-PK1 cells,

RT-PCR was performed using primers corresponding to ALK2 (also known as

Acvr1 and ActRI), a type I receptor that transduces signals from BMP-7 [Macías-

Silva et al., 1998] and is expressed in native proximal tubule cells [Gould et al.,

2003]. Semi-quantitative RT-PCR (Figure 5.2) showed the existence of this receptor and suggested that ATP depletion and recovery induced higher expression of ALK2 mRNA (an increase of 85 + 18%; n=4). To confirm this

71 finding at the protein level, Western analysis was performed on LLC-PK1 cells subjected to various times of ATP depletion/recovery. Figure 5.2 demonstrates similarly increased levels of ALK2 protein in stressed and recovered cells compared with those under normal growth conditions. This finding provides a mechanism by which ATP-depleted/recovered cells can amplify their responsiveness to BMP-7, thus inducing signaling through the Smad 1/5/8 pathway.

5.2.3 Regulatory Mechanisms Mediating Translational Control of HuR

We have shown that HuR gene transcription results in two alternate isoforms of mRNA that are the outcome of precise regulation to avoid deleterious affects of aberrant HuR production [Ayupova and Singh et al., 2009]. These isoforms contain vastly different 5’ UTRs and demonstrate markedly different levels of translatability in in vitro assays. Therefore, it was important to examine the mechanisms of translational control of these forms under differing conditions.

In the following sections, the effects of normal growth, stress, and recovery on

HuR translation will be explored.

5.2.4 Translatability of Alternate Forms of HuR mRNA under Normal,

Stressed, and Recovered Conditions

To determine under which cellular conditions the alternate forms of HuR mRNA are translated, LLC-PK1 cells were cultured normally or subjected to ATP

depletion and recovery. Ribosome fractionation was performed after lysate

72 collection via sucrose density gradient. HuR mRNA and protein were isolated

from these fractions. Using RT-PCR, the presence of these two different forms of

HuR mRNA was detected using different primer sets. One set was specific for

the mRNA1 (long form); the other detected total HuR mRNA, as we cannot

detect mRNA2 (short form) on its own as it is essentially a truncated version of

mRNA1. RT-PCR on the ribosomal fractions showed that total HuR mRNA is

distributed evenly among lighter (cytosolic, monosome) and heavier (polysomes)

fractions (Fig 5.3A). In contrast, mRNA1 is associated with polysomes only

during normal growth and recovery (Fig 5.3B) but was absent from polysomes

during ATP depletion. This finding suggests that mRNA1, and possibly mRNA2,

is capable of being translated during normal growth and recovery, while only

mRNA2 is translated during ATP depletion. This result is consistent with the

hypothesis that mRNA2 is a readily translatable form, and therefore is upregulated following an initial ATP depletion event as a preconditioning

mechanism.

5.2.5 Stress Granules

The ribosome fractionation results above demonstrate that the short form of

HuR mRNA is translated during ATP depletion, whereas the long form is

translated under normal and recovered conditions. This finding suggests that the

transcripts are inactive under different cellular conditions. We hypothesize that

the active transcripts are synthesized by potential partitioning into privileged

translation sites (i.e. polyribosomes), whereas during ATP depletion, the inactive

73 form (i.e. mRNA1) may be sequestered into stress granules (SGs), which are

aggregates of stalled translation initiation complexes that form under various

types of cellular stress. However, nothing is known about the formation of SGs in

renal proximal tubule cells. Therefore, we explored their formation under conditions of ATP depletion and recovery using markers known to accumulate in

SGs. Initial identification of SGs in proximal tubule cells was accomplished

through immunocytochemically labeling the related RNA-binding proteins TIA-1

and TIAR (Kedersha et al., 1999). Under normal conditions TIA-1 and TIAR are

concentrated in the nuclei of cells, but in response to stress, both proteins rapidly

accumulate in the cytoplasm and become robust markers of SGs.

5.2.6 Formation of Stress Granules during ATP Depletion and Recovery

To determine whether ATP depletion and recovery induces SG formation in

kidney epithelial cells, LLC-PK1 cells were ATP depleted, recovered in normal

growth medium, and subjected to immunocytochemical analysis. Discrete foci

became apparent within 30 minutes of ATP depletion, as assayed by immuno-

staining for the SG markers TIAR. These granules become larger and more

apparent over 3-4 hours of recovery (Figure 5.4). Similar results were obtained

when cells were labeled for the related marker TIA-1 (data not shown). These

data are suggestive that ATP depletion of renal epithelia supports stress granule

formation, although further characterization of these foci is required.

74 5.2.7 Effects of Cycloheximide and Puromycin on Stress Granule Formation

in LLC-PK1 Cells

The paradoxically antagonistic effects of different pharmacological inhibitors

of protein translation on SG assembly reveal the dynamic equilibrium between

polysomes and SGs. Drugs that stabilize polysomes by freezing ribosomes on

translating mRNAs (e.g. cycloheximide and emetine) inhibit the assembly of SGs

and actively dissolve them in the continued presence of both stress and eIF2α

phosphorylation (Kedersha et al., 2002). In contrast, drugs that destabilize

polysomes by releasing ribosomes from mRNA transcripts (e.g. puromycin)

promote the assembly of SGs (Kedersha et al., 2002). There is a dynamic equilibrium between polysomes and SGs, which has led investigators to propose that SGs are the sites of mRNA triage, where untranslated mRNAs accumulate during stress prior to degradation, reinitiation, or repackaging as mRNPs

(Kedersha et al., 2002).

To determine whether discrete foci identified during stress and recovery of

LLC-PK1 cells were indeed SGs, cells were ATP depleted/recovered along with

the addition of cyclohexmide or puromycin. Upon puromycin treatment these foci

became larger whereas cycloheximide treatment made them disappear, as

assayed by immunolabeling for the SG marker TIAR (Figure 5.5). These results

are consistent with the identification of these foci as stress granules.

5.2.8 Distribution of Stress Granule Markers during ATP Depletion and

Recovery

75 Stress granules are primarily composed of stalled 48S complexes

containing bound mRNAs derived from disassembling polysomes. These contain

poly(A) mRNA bound to early initiation factors (such as eIF4E, eIF3, eIF4A,

eIFG) and small, but not large, ribosomal subunits. To further characterize the

putative stress granules seen in our cell model, large ribosomal proteins and

eIF2-alpha, which are known to be absent from SGs, were tested for their presence or absence in the stress granules formed in our ATP depletion model.

Using immunocytochemical methods, we tested for the presence of eIF2α and the large ribosomal protein, Ribo-P antigen. As shown in Figure 5.6, their distributions showed no overlap with that of the SG foci. In addition, we used antibodies against individual components of the 48S pre-initiation complex (e.g. eIF3, eIF4E, and PABP) to determine whether these proteins were present in the

SG foci. Most components of the 48S pre-initiation complex were found to colocalize with TIAR at the foci (Figure 5.7). These results indicate that in LLC-

PK1 cells, the discrete foci represent SGs that are eIF2-eIF5–deficient, pre-

initiation complexes that are assembled during stress.

RNA-binding proteins that either promote [Gallouzi et al., 2000] or inhibit

[Kedersha et al., 2000] mRNA stability are also recruited to SGs. This suggests

that the SG is a site where the fates of specific transcripts are determined by the

activity of different RNA-binding proteins. Consistent with other stress models,

poly(A) mRNA was found to be present in stress granules induced by ATP

depletion (Figure 5.7), as assessed by probing cells with a fluorescent oligo-dT

76 oligonucleotide. This finding is consistent with previous studies showing that poly(A) mRNA is a component of mammalian SGs [Kedersha et al., 1999].

5.2.9 Increase in the Phosphorylation Level of eIF2-alpha during ATP

Depletion

Eukaryotic cells express a family of translation initiation factor 2 alpha

(eIF2α) kinases (PKR, PERK, GCN2, HRI) that are individually activated in response to distinct types of environmental stress. Phosphorylation of eIF2α by one or more of these kinases reduces the concentration of eIF2–guanosine triphosphate (GTP)–transfer ribonucleic acid for methionine (tRNA-Met), the ternary complex that loads tRNA-Met onto the small ribosomal subunit to initiate protein translation. When ternary complex levels are reduced, the related RNA- binding proteins TIA-1 and TIAR promote the assembly of a noncanonical pre- initiation complex that lacks eIF2-GTP-tRNA-Met. The TIA proteins dynamically sort these translationally incompetent pre-initiation complexes into stress granules.

It has been reported in certain models that eIF2-alpha phosphorylation increases upon stress. This phosphorylation event is required to suppress general protein translation during stress. However, it has been suggested that

ATP depletion may inhibit translation by an alternate mechanism [Kedersha et al., 2002]. In order to determine the phosphorylation status of eIF2α in energy- depleted proximal tubules, LLC-PK1 cells were subjected to stress by ATP depletion and the phosphorylation state of eIF2α was determined by Western

77 blot. Phosphorylation of eIF2α increased during ATP depletion (Figure 5.8A) and

decreased during recovered conditions (Figure 5.8B), while total eIF2α remained unchanged. This finding suggests that general protein translation is diminished during ATP depletion. However, as we previously demonstrated, translation of

HuR (the short mRNA isoform) continues and indeed increases during ATP depletion. Thus, translation of this mRNA isoform appears to be able to circumvent the normal suppressive effects of eIF2-alpha phosphorylation.

5.2.10 PERK is Activated by ATP Depletion of Proximal Tubule Cells

The eIF2α subunit is the target of a family of serine/threonine kinases (PKR,

PERK, GCN2, HRI) that are activated by different forms of environmental stress.

For example, PKR senses heat, ultraviolet (UV) irradiation, viral infection, and

oxidative stress [Williams, 2001], whereas PERK (also known as PEK) detects

endoplasmic reticulum stress. GCN2 senses amino acid starvation [Kimball,

2001], and HRI monitors changes in the availability of heme during erythrocyte

differentiation [Han et al., 2001; Lu et al., 2001]. Each of these stress-activated kinases phosphorylates eIF2α on serine 51, a modification that increases the affinity of eIF2 for eIF2B, a GDP-GTP exchange factor that charges the eIF2-

GTP-tRNA-Met ternary complex [Kimball, 2001].

Endoplasmic reticulum (ER) stress, which can lead to activation of the eIF2

kinase PERK and eIF2α phosphorylation, has been shown to accompany

ischemic injury in several tissues, including kidney epithelia [Kumar et al., 2001].

Translational repression from ER stress has long been known to be a transient

78 phenomenon [Brostrom et al., 1998; Kaufman, 1999], and phosphorylation of

eIF2α by endoplasmic reticulum (ER) stress, which can lead to activation of the

eIF2 kinase PERK and eIF2α phosphorylation, has been shown to accompany ischemic injury in several tissues, including kidney epithelia [Bush et al., 2000].

More recently, renal ischemia-reperfusion was shown to induce phosphorylation of eIF2α via PERK directly [Montie et al., 2005].

Because PERK activity has been implicated in renal stress caused by ischemia-reperfusion, the phosphorylation status of PERK was determined by

Western analysis during ATP depletion of LLC-PK1 cells. It was observed that

phosphorylation of PERK increased within 15 minutes of incubation of cells in

ATP depletion buffer, while total PERK levels remained constant (Figure 5.8C).

Although this experiment does not rule out involvement of the other eIF2-alpha

kinases, it indicates a likelihood that PERK plays a key role in phosphorylation of eIF2-alpha and subsequent stress granule formation and translational repression in our model system.

5.2.11 Post-transcriptional Control Element (PCEs) and Translation of HuR

As described earlier in this chapter, one isoform of HuR mRNA (mRNA1)

contains a long, highly structured 5’ UTR. This mRNA was shown to be poorly

translated in rabbit reticulocyte lysates [Ayupova and Singh et al., 2009]. This

finding begs the question, what is the purpose of synthesizing a poorly translated

mRNA? One possibility is that this mRNA contains an internal ribosome entry

site (IRES). IRESs are elements found in highly structured 5’ UTRs that help

79 initiate ribosome binding near the initiation codon, rather than at the mRNA 5’ cap [Pestova et al., 2001]. This circumvents the need for ribosomal complexes to scan through a highly structured 5’ UTR. The possibility of HuR mRNA1

containing an IRES was highly attractive, as IRESs are often used during times

of cellular stress [Kullmann et al., 2003; Meng et al., 2005]. However, other work

in the Lee laboratory demonstrated an absence of IRES activity in the 5’ UTR of

HuR mRNA1 (data not shown). Therefore, it seemed likely that an alternate form

of initiation might be required to translate this mRNA.

Recently, another form of translation initiation was identified in retroviruses

and a few mammalian mRNAs, termed the post-transcriptional control element

(PCE). PCEs are extensively studied by the laboratory of Dr. Kathleen Boris-

Lawrie of the OSU College of Veterinary Medicine. PCEs are highly structured 5’

UTR sequences that facilitate, rather than inhibit, translation initiation. PCEs are

bound by RNA helicase A (RHA), which further recruits ribosomal complexes and

aids in unwinding the secondary structure of the 5’ UTR [Gritta et al., 2006],

allowing cap-dependent translation. PCEs form a tripartite stem-loop structure

composed of three required structural motifs. The HuR mRNA1 5’ UTR was

compared to the 5’ UTR of transcription factor JunD, which has been established

to contain a PCE [Hernandez et al., 2008]. Figure 5.9A shows high between HuR and JunD within JunD’s established structural motifs.

Further, the laboratory of Boris-Lawrie, using a PCE reporter assay developed in

their group, found that HuR mRNA1 exhibits activity similar to that of a known

PCE-containing retrovirus (Figure 5.9B). Finally, in order to determine the

80 potential role of RNA helicase A (RHA) in regulating expression of HuR, we

knocked down expression of RHA using siRNAs developed by the Boris-Lawrie

lab. This knockdown in proximal tubule cells resulted in suppression of HuR

protein levels (Figure 5.9C). Taken together, these data strongly suggest the presence of a PCE in the 5’ UTR of HuR mRNA1.

5.3 CONCLUSIONS

Human antigen R (HuR) is an RNA-binding protein with protective activities

against cellular stress. Under basal conditions, HuR mRNA is expressed in two

forms: one that contains a 20-base 5’-untranslated region (UTR) sequence and

one that contains a 150-base, GC-rich 5’-UTR that is inhibitory to translation in

vitro. Previous results showed that recovery from cellular stresses such as

thapsigargin and ATP depletion induced increased expression of the shorter,

more translatable transcript (mRNA2) and decreased expression of the longer

form (mRNA1). Analysis of HuR upstream regions revealed sequences

necessary for regulation of the shorter mRNA that exist within the exon encoding

the GC-rich 5’-UTR of the longer mRNA. Gel shift and chromatin

immunoprecipitation analyses demonstrated the ability of Smad 1/5/8

transcription factors to bind to these sequences that contain overlapping copies

of the motif GCCGnCGC. Because Smad 1/5/8 are induced by bone

morphogenetic proteins (BMPs), these results suggest a role for HuR in

regulating the pleiotropic effects of BMP-7 in recovery from renal stress.

81 Because HuR mRNA isoforms contain different 5’ UTRs and have shown

different levels of translatability in in vitro assays, the effects of normal growth, stress, and recovery on HuR translation were explored. HuR mRNA1 was shown to contain a PCE, which imposes a translational regulation and affects the levels of HuR expression during normal growth and recovered conditions. However, mRNA1 was not found to be translated during the stress of ATP depletion. Given these findings, as well as the presence of stress granules in ATP-depleted proximal tubule cells, we hypothesize that differential translation of the HuR mRNAs is likely to be a function of mRNA sequestration to stress granules or polyribosomes.

82

A

B

Figure 5.1. Smad 1/5/8 Binding Sites are Present in the HuR Promoter.

(A) LLC-PK1 cells were cultured in normal growth medium or ATP depletion medium for 2 hours and allowed to recover. Gel mobility shift assays were performed on nuclear extracts from untreated (unt) or ATP-depleted/ recovered

(rec) LLC-PK1 cells. One major band (designated by the arrow) was found to specifically bind Smad 1/5/8 consensus motifs in the porcine HuR 5’-UTR. The intensity of this band increased following ATP depletion/recovery. (B) In chromatin immunoprecipitation (ChIP) assays, antibody against Smad 1/5/8 precipitated DNA corresponding to the HuR 5’-UTR, as detected by PCR using specific primers. Although levels of input DNA were unchanged, Smad 1/5/8 antibodies precipitated more HuR DNA from ATP depleted/recovered cells than untreated cells, consistent with the gel mobility shift findings.

83

Figure 5.2. ATP Depletion and Recovery Induces Expression of ALK2. Two hours of ATP depletion followed by up to 4 hours of recovery caused increased expression of ALK2 mRNA (left panel) and protein (right panel). A beta-actin immunoblot is included as a loading control.

84

Figure 5.3. Ribosomal Fractionation Indicates Differential Translation of HuR mRNAs during Normal Growth, ATP Depletion, and Recovery. Ribosomal fractionation shows that total HuR mRNA is distributed across all the fractions (left panel), whereas mRNA1 (the long isoform) is heavily associated with polysomes during normal growth and recovery from ATP depletion (right panel). In contrast, mRNA2 (the short isoform) alone is translated during ATP depletion.

85

Figure 5.4. ATP Depletion Induces Stress Granule Formation. LLC-PK1 cells were plated on glass coverslips and were treated with D-glucose free media and 0.1 µM antimycin A for 1 to 4 hours followed by recovery in normal growth media for up to 4 hours. Cells were fixed, permeabilized, and immunolabeled with antibodies against TIAR.

86

Figure 5.5. Inhibitors of Translation Alter Stress Granule Formation in ATP- depleted LLC-PK1 cells. LLC-PK1 cells were plated on glass coverslips and were ATP depleted for 1 hour followed by 2 or 4 hours of recovery in media containing 10 µg/ml puromycin or 100 µg/ml cycloheximide. Cells were fixed, permeabilized, and immunolabeled with antibodies against TIAR.

87

Figure 5.6. Neither eIF2-alpha nor a Large Ribosomal Subunit Co-Localize

with Stress Granules in LLC-PK1 Cells. LLC-PK1cells were plated on glass coverslips and were ATP depleted for 1 hour followed by 2 or 4 hours of recovery in normal growth media. (top row) Cells were fixed, permeabilized, and immunolabeled with antibodies against TIAR in green and eIF2α in red. (bottom row) Cells were labeled with antibodies against large ribosomal subunit Ribo-P Antigen (green) and TIAR (red).

88

Figure 5.7. Translation Initiation Factors, Small Ribosomal Proteins, and poly(A) mRNA Segregate to Stress Granules in LLC-PK1 Cells. Initiation factors such as eIF2αP, eIF3, with TIAR show their presence in SGs along with small ribosomal protein S6 and poly(A) mRNA. HuR protein was also localized to SGs. Cells were fixed, permeabilized, immunostained with antibodies against phospho-eIF2-alpha, eIF3 and S6 in red, and HuR in green. Poly(A) mRNA was detected using a fluorescent labeled oligo-dT probe.

89

A

B

C

Figure 5.8. ATP Depletion Induces Phosphorylation of eIF2-alpha and

PERK. (A) LLC-PK1 cells were ATP depleted for up to 4 hours and lysates were assessed by Western blot for levels of total and phosphorylated eIF2-alpha. (B)

LLC-PK1 cells were ATP depleted 1 hour and recovered in normal medium to up to 4 hours. Lysates were assessed for levels of total and phosphorylated eIF2-

alpha as in (A). (C) LLC-PK1 cells were ATP depleted for up to 30 minutes and assessed by Western blot for levels of total and phosphorylated PERK.

90

Figure 5.9. HuR mRNA Contains a Post-transcriptional Control Element. (A) PCE-like motifs in HuR mRNA1 shows high homology with the PCE elements in JunD. (B) The HuR mRNA1 5’ UTR shows comparable PCE activity to that of SNV PCE, assayed via reporter constructs. (C) siRNA mediated knockdown of RHA results in suppression of HuR protein levels as assessed by Western blotting. β-actin is included as a loading control.

91

CHAPTER 6

Discussion

6.1 Anti-apoptotic Role of HuR

HuR, as one of the regulators of mRNA stability in renal epithelial cells, is redistributed and regulated at multiple levels during energy depletion and recovery. By binding to mRNAs containing specific AREs, HuR contributes to the stability of hundreds to thousands of distinct mRNA transcripts. It is a stress response protein that is known to play an important role in cell survival, as its loss in vitro or in vivo results in heightened apoptosis [Wang et al., 2000]. By stabilizing and promoting translation of specific mRNAs during stress events,

HuR is protective against cellular damage. It binds to mRNAs that encode several major categories of proteins including modulators of cell proliferation and differentiation, inflammation, cell survival and apoptosis. Because HuR appears to play a role in stabilizing numerous transcripts during cellular stress and apoptosis [Wang et al., 2000], understanding the regulation of this protein gives an important insight into general mechanisms of survival during environmental insults such as ischemic injury.

Although HuR has the capacity to bind numerous mRNA targets, it is known to promote mRNA stabilization or translation of a few specific gene products that

92 play important roles in promoting cell proliferation or protecting against apoptosis

and senescence. Target mRNA ligands for HuR include cell cycle-associated gene products such as c-Fos, p21, p53, and various cyclins, anti-apoptotic proteins including prothymosin α, and SIRT1, and other stress-related proteins such as HIF1α. HuR has emerged as an elicitor of a broad antiapoptotic function through its influence on the expression of proteins directly (e.g., ProTα, SIRT,

Bcl-2, Mcl-1) or indirectly (e.g., p21, p27, EGF, VEGF, IGF-IR), preventing apoptosis [Lopez et al., 2005].

HuR, together with polypyrimidine binding protein, binds hypoxia-inducible

factor-1 α (HIF-1α) mRNA and promotes HIF-1α translation under hypoxia-like

conditions [Galban et al., 2008]. Activation of HIF-1α has been identified as an important mechanism of cellular adaptation to low oxygen, one of the conditions arising from ischemia at the cellular level. Under a decreased oxygen level, HIF-

1α regulates a broad range of physiologically relevant genes involved in erythropoiesis, angiogenesis, apoptosis, and energy metabolism [Maxwell, 2003].

It was shown that preconditional activation of HIF-1α protected against ischemic injury in rats [Bernhardt, 2006]. It is striking that many of HIF-1 α target genes are also regulated posttranscriptionally by HuR, including VEGF, COX-2, IL-6, IL-8, and GLUT1 [Gantt et al., 2006; Mrena et al., 2005], and likely further contribute to the protective and adaptive role of HuR under ischemic conditions. By acting upon various mRNA pools, HuR participates in numerous key cellular processes; thus it was of interest to know the mechanism of how HuR expression is regulated.

93 Our own studies have demonstrated the anti-apoptotic nature of HuR in

cultured renal proximal tubule cells subjected to ATP depletion and indicate Bcl-2

and Hsp70 as targets [Ayupova and Singh et al., 2009]. Therefore, the

expression of HuR in kidney epithelia subjected to ischemia/reperfusion injury

may be protective against apoptosis caused by the ischemic insult and may also

promote proliferation of newly regenerating proximal tubule epithelia.

6.2 HuR, PI3 Kinase/Akt Pathway, and Apoptosis

We demonstrated that a major effect of HuR knockdown in cultured

proximal tubule cells was suppression of anti-apoptotic mRNAs including those

within the Akt signaling pathway. Other mRNAs that were downregulated in HuR-

suppressed cells (not discussed above) were transcripts corresponding to anti-

apoptotic proteins including the Bcl-2 family, Mcl-1, NAIP, NOD-1, and BFAR.

We found that HuR plays a central role in PI3K/Akt signaling, as it enhances

expression of Grb10, which promotes activation of Akt, that further goes on to

upregulated HuR expression. Thus, HuR is involved in a positive feedback loop

promoting Akt activity and cell survival. This study demonstrates how HuR plays

a central, positive role in proximal tubule cell survival through the activities of Akt.

As described above, HuR has been demonstrated to regulate the mRNA stability and/or translation of critical cell survival proteins in addition to Grb10.

HuR’s involvement in PI3K/Akt signaling adds another layer of complexity to its

effects on cell survival. For example, expression of the caspase inhibitor XIAP (X

-linked inhibitor of apoptosis) is increased by HuR both at the level

94 of mRNA stability (Zhang et al., 2009) and translation (Durie et al., 2010). Here we show that HuR can act on XIAP at another level to enhance cell survival; that is, Akt phosphorylates XIAP, inhibiting its ubiquitination and degradation. Thus,

HuR directly interacts with XIAP mRNA to increase its expression, while Akt, which is indirectly activated by HuR through Grb10, increases XIAP’s expression post-translationally. In another example, HuR also has been demonstrated to increase Bcl-2 expression at the levels of both mRNA stability and translation

(Ishimaru et al., 2009). Bcl-2’s ability to enhance cell survival lies in part in its abundance relative to that of its pro-apoptotic family member BAD, which when

dimerized to Bcl-2 (or Bcl-XL), promotes apoptosis. Akt phosphorylates BAD, resulting in its dissociation from Bcl-2/Bcl-XL and association with 14-3-3, thus inhibiting apoptosis. Therefore, while HuR increases Bcl-2 expression by

interaction with Bcl-2 mRNA, it also increases Bcl-2 activity through Akt-mediated

degradation of BAD.

The ability of HuR to affect anti-apoptotic pathways at multiple levels

accounts for its importance in promoting cell survival. Knockout of HuR pre-

natally results in embryonic lethality [Katsanou et al., 2009], while knockout post-

natally results in rapid death [Ghosh et al., 2009]. Conversely, overexpression of

HuR by as little as three to four-fold has been associated with increased

tumorigenicity in numerous tissue types, including renal cell carcinomas [Danilin

et al., 2010]. For this reason, HuR expression must be tightly controlled.

We have identified the adapter protein Grb10 as a new target of HuR

regulation and have shown its importance in mediating cell survival during stress.

95 As briefly described above, Grb10 has been demonstrated to bind Akt and

transport it to the plasma membrane where this protein complex may associate

with various activated receptor tyrosine kinases through Grb10’s SH2 domain

[Jahn et al., 2002]. This association brings Akt in proximity to plasma membrane

PI3 kinase, resulting in Akt’s phosphorylation and activation. Additionally, other

studies have indicated that Grb10 can promote cell survival through MAP kinase

pathways as well as Akt [Kebache et al., 2007]. Interestingly, HK-2 cells under

normal conditions demonstrate a primarily nuclear localization of pAkt. While in

the nucleus, pAkt has access to its nuclear targets including the forkhead family

of transcription factors. However, ATP depletion results in pAkt movement to the

plasma membrane, where it may have access to a new set of targets. Grb10 is

required both for pAkt activation and its movement to the plasma membrane.

Although our studies and other indicate that Grb10 promotes cell survival, it

is a negative regulator of growth, as its disruption in mice leads to embryonic and

placental overgrowth [Charalambous et al., 2003]. This is most likely due to

inhibition of insulin and insulin-like growth factor receptor signaling. Interestingly,

HuR also has been noted to inhibit insulin-like growth factor receptor signaling,

through inhibition of translation of the type I receptor [Meng et al., 2005].

However, it must be noted that the Grb10 literature contains multiple examples of

this adapter playing both negative and positive roles in studies of insulin signaling in vitro [Lim et al., 2004; Riedel, 2004]. These discrepancies may be attributable

in part to tissue- and cell-specific differences or to artifacts associated with

overexpression of Grb10 protein. Grb10 mRNA is expressed as multiple

96 alternately spliced transcripts (3 in mice, 4 in humans) whose functions may or

may not differ. In the present studies, we used Grb10 siRNAs that knocked down

expression of all transcripts so that any isoform-specific effects would not occur,

and our RT-PCR primers and antibodies also detected all forms. Further, in our studies to determine the role of the Grb10 3’ UTR in its expression, we transfected into HK-2 cells a mouse Grb10eta cDNA. We expect the findings

from this experiment to be relevant to all murine Grb10 isoforms, as they contain

identical 3’ untranslated regions.

In our study, Grb10 has emerged a novel binding target of HuR, which

plays a key role in regulating the PI3K/Akt pathway for cell survival. Therefore,

HuR demonstrates a central role in cell survival as both an activator of PI3K/Akt signaling and as a downstream target of PI3K/Akt-mediated gene regulation in renal epithelia.

6.3 Transcriptional and Translational Control of HuR

We previously demonstrated that HuR mRNA is synthesized in two forms

that differ in their 5’ untranslated regions and thus are transcribed through the

action of alternative promoters [Ayupova and Singh et al., 2009]. The data

presented here provide an initial characterization of essential promoter elements

required for transcriptional activation of HuR mRNA2 during recovery of renal

epithelia from cellular stress. Characterization of promoter elements driving

transcription of an mRNA1 is underway. Genome wide analysis has revealed the

prevalence of numerous alternate promoters, in fact suggesting that up to half of

97 human genes produce variant transcripts [Tsuritani et al., 2007]. Transcription from alternate promoters has been shown to be induced under multiple circumstances, including developmental activity [Saleh et al., 2002] tissue specificity [kamat et al., 2002], and stress [Meshorer et al., 2004]. Further, alternative promoter transcripts previously have been shown effective in the control of translational machinery, as suggested here for HuR [Landry et al.,

2003].

Because HuR mRNA is present as two isoforms contain different 5’ UTRs

and have shown different levels of translatability in in vitro assays, the effects of

normal growth, stress, and recovery on HuR translation was explored. Although

the HuR mRNA bearing the short 5’-UTR (mRNA2) is under control of

BMP/Smad signaling and is readily translatable, the mechanisms by which the

longer transcript (mRNA1) is translated were not known. Our results indicate that

both isoforms of HuR mRNA are present during stressed conditions; however

mRNA1 was found not to be translated during this time. Interestingly, the 5’ UTR

of mRNA1 was shown to contain a post-transcriptional control element (PCE).

The PCE imposes a translational regulation and affects the levels of HuR

expression during normal growth and recovered conditions. Although the role of

PCEs in mammalian cells is not understood, we speculate that the presence of a

PCE in mRNA1 aids to keep a check on the translation levels of HuR during

normal and recovered conditions. However, when a cell is exposed to and

recovers from stress, synthesize of mRNA2 predominates so that during

repeated stresses, this more translatable transcript may be readily translated.

98 We also observed that cultured proximal tubule cells subjected to ATP depletion form the foci of stalled translation initiation known as stress granules.

Although translation may be inhibited under numerous types of stresses, not all of these induce SG formation. Here we found that ATP depletion does indeed cause formation of these aggregates, and that phosphorylation of eIF2-alpha by the stress kinase PERK is likely to play a key role in this process. We also determined that some HuR mRNA is associated with SGs during ATP depletion.

However, we have been yet unable to determine which mRNA isoform or isoforms is present in these granules. Because mRNA1 is not translated during

ATP depletion, we speculate this is due to its sequestration in SGs. This is further bolstered by the findings that RNA helicase A, the PCE binding protein, is present in SGs during oxidative stress brought about by arsenite exposure (Dr.

K. Boris-Lawrie et al., unpublished data). This finding is consistent with the PCE- containing mRNA1 also being sequestered in SGs during ATP depletion.

However, more experiments are required to definitively prove the identity of the

SG-associated transcript(s). Nonetheless, observing these findings, it is hypothesized that differential translation of the HuR mRNAs is likely to be a function of alternative mRNA targeting to stress granules or polyribosomes. In summary, it is proposed that modulating the expression of HuR during ischemia/reperfusion injury provides a system in which there are multiple levels of regulation working in concert to protect and restore the cell.

99

BIBLIOGRAPHY

Abdelmohsen K, Lal A, Kim HH, Gorospe M. Posttranscriptional orchestration of an anti-apoptotic program by HuR. Cell Cycle 6: 1288–1292, 2007. Abdelmohsen K, Pullmann R Jr, Lal A, Kim HH, Galban S, Yang X, Blethrow JD, Walker M, Shubert J, Gillespie DA, Furneaux H, Gorospe M. Phosphorylation of HuR by Chk2 regulates SIRT1 expression. Mol Cell 25: 543–557, 2007. Aguila HL, Michaud J, Ai Y, Wu MT, Hemmes A, Ristimaki A, Guo C, Furneaux H, Hla T. Essential role of the RNA-binding protein HuR in progenitor cell survival in mice. J Clin Invest 119:3530-43, 2009. Amsler, K. Role of cell density/cell-cell contact, and growth state in expression of

differentiated properties by the LLC-PK1 cell. J Cell Physiol 159(2): 331-9, 1994. Amsler K, Cook JS. Development of Na+-dependent hexose transport in a cultured line of porcine kidney cells. Am J Physiol 242(1): C94-101, 1982. Anderson P, Kedersha N: Stressful initiations. J Cell Sci 115:3227-3234, 2002. Andreucci M, Michael A, Kramers C, Park KM, Chen A, Matthaeus T, Alessandrini A , Haq S, Force T, Bonventre JV. Renal ischemia/reperfusion and ATP depletion/repletion in LLC-PK1 cells result in phosphorylation of FKHR and FKHRL1. Kidney Int 64:1189–1198, 2003. Ashkenazi A. Targeting death and decoy receptors of the tumour-necrosis factor superfamily. Nat Rev Cancer 2(6): 420-30, 2002. Atasoy U, Watson J, Patel D, Keene JD. ELAV protein HuA (HuR) can redistribute between nucleus and cytoplasm and is upregulated during serum stimulation and T cell activation. J Cell Sci 111 ( 21):3145-56, 1998. Ayupova DA, Singh M, Leonard EC, Basile DP, Lee BS. Expression of the RNA- stabilizing protein HuR in ischemia-reperfusion injury of rat kidney. Am J Physiol Renal Physiol 297:95-105, 2009. Bakheet T, Williams BR, Khabar KS. ARED 3.0: the large and diverse AU-rich transcriptome. Nucleic Acids Res 34:111-4, 2006. Bashirullah A, Cooperstock RL, Lipshitz HD. Spatial and temporal control of RNA stability. Proc Natl Acad Sci USA 98:7025-7028, 2001. Basile DP, Liapis H, Hammerman MR. Expression of bcl-2 and bax in regenerating rat renal tubules following ischemic injury. Am J Physiol Renal Physiol 272: 640–647, 1997. 100 Bastin J, Cambon N, Thompson M, Lowry OH, and Burch HB. Change in energy reserves in different segments of the nephron during brief ischemia. Kidney Int 31:1239-1247, 1987. Becker LB. New concepts in reactive oxygen species and cardiovascular reperfusion physiology. Cardiovasc Res 61:461–70, 2004. Bernhardt WM, Campean V, Kany S, Jurgensen JS, Weidemann A, Warnecke C, Arend M, Klaus S, Gunzler V, Amann K, Willam C, Wiesener MS, Eckardt KU. Preconditional activation of hypoxia-inducible factors ameliorates ischemic acute renal failure. J Am Soc Nephrol 17: 1970–1978, 2006. Bidmon B, Endemann M, Müller T, Arbeiter K, Herkner K, Aufricht C. Heat shock protein-70 repairs proximal tubule structure after renal ischemia. Kidney Int 58: 2400–2407, 2000. Boris-Lawrie K, Temin HM. Genetically simpler bovine leukemia virus derivatives can replicate independently of Tax and Rex. J Virol 69:1920-1924, 1995. Brennan CM, Steitz JA. HuR and mRNA stability. Cell Mol Life Sci 58: 266–277, 2001. Brostrom CO, Brostrom MA. Regulation of translational initiation during cellular responses to stress. Prog Nucleic Acid Res Mol Biol 58: 79-125, 1998. Bush KT, Keller SH, Nigam SK. Genesis and reversal of the ischemic phenotype in epithelial cells. J Clin Invest 106(5): 621-6, 2000. Canfield PE, Geerdes AM, Molitoris BA. Effect of reversible ATP depletion on

tight-junction integrity in LLC-PK1 cells. Am J Physiol 261:1038-1045, 1991. Castaneda MP, Swiatecka-Urban A, Mitsnefes MM, Feuerstein D, Kaskel FJ, Tellis V, Devarajan P. Activation of mitochondrial apoptotic pathways in human renal allografts following ischemia-reperfusion. Transplantation 76: 50–54, 2003. Charalambous M, Smith FM, Bennett WR, Crew TE, Mackenzie F, Ward A. Disruption of the imprinted Grb10 gene leads to disproportionate overgrowth by an Igf2-independent mechanism. Proc Natl Acad Sci USA. 100:8292-7, 2003. Chen H, Xing B, Liu X, Zhan B, Zhou J, Zhu H, Chen Z. Ischemic postconditioning inhibits apoptosis after renal ischemia/reperfusion injury in rat. Transpl Int 21:364-71, 2008. Chiang-Ting C, Tzu-Ching C, Ching-Yi T, Song-Kuen S, Ming-Kuen L. Adenovirus-mediated bcl-2 gene transfer inhibits renal ischemia/reperfusion induced tubular oxidative stress and apoptosis. Am J Transplant 5: 1194– 1203, 2005. Chien CT, Shyue SK, Lai MK. Bcl-xL augmentation potentially reduces ischemia/reperfusion induced proximal and distal tubular apoptosis and autophagy. Transplantation 84: 1183–1190, 2007.

101 Conne B, Stutz A, Vassalli JD. The 3' untranslated region of messenger RNA: a molecular 'hotspot' for pathology? Nat Med 6:637-641, 2000. Dancey JE. Molecular targeting: PI3 kinase pathway. Annals Oncol 15 (4): 233– 239, 2004. Danilin S, Sourbier C, Thomas L, Lindner V, Rothhut S, Dormoy V, Helwig JJ, Jacqmin D, Lang H, Massfelder T. Role of the RNA-binding protein HuR in human renal cell carcinoma. Carcinogenesis. 31:1018-26, 2010. Datta SR, Brunet A, Greenberg ME. Cellular survival: a play in three Akts. Genes Dev 13: 2905-2927, 1999. Denkert C, Weichert W, Pest S, Koch I, Licht D, Kobel M, Reles A, Sehouli J, Dietel M, Hauptmann S. Overexpression of the embryonic-lethal abnormal vision-like protein HuR in ovarian carcinoma is a prognostic factor and is associated with increased cyclooxygenase 2 expression. Cancer Res 64:189-95, 2004. Denkert C, Weichert W, Winzer KJ, Muller BM, Noske A, Niesporek S, Kristiansen G, Guski H, Dietel M, Hauptmann S. Expression of the ELAV- like protein HuR is associated with higher tumor grade and increased cyclooxygenase-2 expression in human breast carcinoma. Clin Cancer Res 10:5580-6, 2004. Devarajan, P. Update on mechanisms of ischemic acute kidney injury. J Am Soc Nephrol 17(6): 1503-20, 2006. Devarajan P, Mishra J, Supavekin S, Patterson LT, Potter S. Gene expression in early ischemic renal injury: clues towards pathogenesis, biomarker discovery, and novel therapeutics. Mol Genetics and Metabol 80: 365–376, 2003. Dudley AT, Lyson KM, Robertson EJ. A requirement for bone morphogenetic protein-7 during development of the mammalian kidney and eye. Genes Dev 9: 2795–2807, 1995. Earnshaw WC, Martins LM, Kaufmann SH. Mammalian caspases: structure, activation, substrates, and functions during apoptosis. Annu Rev Biochem 68: 383-424, 1999. Fan XC, Steitz JA. Overexpression of HuR, a nuclear-cytoplasmic shuttling protein, increases the in vivo stability of ARE-containing mRNAs. EMBO J 17:3448-3460, 1998. Galban S, Kuwano Y, Pullmann R Jr, Martindale JL, Kim HH, Lal A, Abdelmohsen K, Yang X, Dang Y, Liu JO, Lewis SM, Holcik M, Gorospe M. RNA-binding proteins HuR and PTB promote the translation of hypoxia- inducible factor 1 alpha. Mol Cell Biol 28: 93–107, 2008.

102 Gallouzi IE, Brennan CM, Steitz JA. Protein ligands mediate the CRM1- dependent export of HuR in response to heat shock. RNA 7: 1348–1361, 2001. Gantt KR, Cherry J, Richardson M, Karschner V, Atasoy U, Pekala PH. The regulation of glucose transporter (GLUT1) expression by the RNA binding protein HuR. J Cell Biochem 99: 565–574, 2006. Gobe G, Willgoss 0, Hogg N, Schoch E, Endre Z. Cell survival or death in renal tubular epithelium after ischemia-reperfusion injury. Kidney Int 56:1299- 1304, 1999. Gould SE, Day M, Jones SS, Dorai H. BMP-7 regulates chemokine, cytokine, and hemodynamic gene expression in proximal tubule cells. Kidney Int 61: 51–60, 2002. Halees AS, El-Badrawi R, Khabar KS. ARED Organism: expansion of ARED reveals AU-rich element cluster variations between human and mouse. Nucleic Acids Res 36:D137-40, 2008. Hartman TR, Qian S, Bolinger C, Fernandez S, Schoenberg DR, Boris-Lawrie K. RNA helicase A is necessary for translation of selected messenger RNAs Nat Struct Mol Biol 13(6):509-16, 2006. Hernandez JM, Floyd DH, Weilbaecher KN, Green PL, Boris-Lawrie K. Multiple facets of junD gene expression are atypical among AP-1 family members. Oncogene 27 (35): 4757-67, 2008. Hruska KA, Guo G, Wozniak M, Martin D, Miller S, Liapis H, Loveday K, Klahr S, Sampath TK, Morrissey. Osteogenic protein-1 prevents renal fibrogenesis associated with ureteral obstruction. Am J Physiol Renal Physiol 279: 130– 143, 2000. Hull S, Boris-Lawrie K. RU5 of Mason-Pfizer Monkey Virus 5' long terminal repeat enhances cytoplasmic expression of human immunodeficiency virus type 1 gag-pol and nonviral reporter RNA. J Virol 76(20): 10211-10218, 2002. Ishimaru D, Ramalingam S, Sengupta TK, Bandyopadhyay S, Dellis S, Tholanikunnel BG, Fernandes DJ, Spicer EK. Regulation of Bcl-2 expression by HuR in HL60 leukemia cells and A431 carcinoma cells. Mol Cancer Res 7:1354-66, 2009. Izquierdo JM. Hu antigen R (HuR) functions as an alternative pre-mRNA splicing regulator of Fas apoptosis-promoting receptor on exon definition. J Biol Chem 283: 19077–19084, 2008. Jahn T, Seipel P, Urschel S, Peschel C, Duyster J. Role for the adaptor protein Grb10 in the activation of Akt. Mol Cell Biol 22:979-91, 2002. Jansen RP. mRNA localization: message on the move. Nat Rev Mol Cell Biol 2:247-256, 2001.

103 Jeyaraj S, Dakhlallah D, Hill SR, Lee BS. HuR stabilizes vacuolar H+ translocating ATPase mRNA during cellular energy depletion. J Bio lChem 280: 37957–37964, 2005. Jeyaraj SC, Dakhlallah D, Hill SR, Lee BS. Expression and distribution of HuR during ATP depletion and recovery in proximal tubule cells. Am J Physiol Renal Physiol 291: 1255–1263, 2006. Jeyaraj SC, Singh M, Ayupova DA, Govindaraju S, Lee BS. Transcriptional control of human antigen R by bone morphogenetic protein. J Biol Chem 285:4432-40, 2010. Joo JD, Kim M, D'Agati VD, Lee HT. Ischemic preconditioning provides both acute and delayed protection against renal ischemia and reperfusion injury in mice. J Am Soc Nephrol 17:3115-23, 2006. Kamat A, Hinshelwood MM, Murry BA, Mendelson, CR. Mechanisms in tissue- specific regulation of estrogen biosynthesis in humans. Trends Endocrinol Metab 13, 122–128, 2002. Kang MJ, Ryu BK, Lee MG, Han J, Lee JH, Ha TK, Byun DS, Chae KS, Lee BH, Chun HS, Lee KY, Kim HJ, Chi SG. NF-kappaB activates transcription of the RNA-binding factor HuR, via PI3K-AKT signaling, to promote gastric tumorigenesis. Gastroenterology 135:2030-42, 2008. Katsanou V, Milatos S, Yiakouvaki A, Sgantzis N, Kotsoni A, Alexiou M, Harokopos V, Aidinis V, Hemberger M, Kontoyiannis DL. The RNA-binding protein Elavl1/HuR is essential for placental branching morphogenesis and embryonic development. Mol Cell Biol 29:2762-76, 2009. Kaufman RJ. Stress signaling from the lumen of the endoplasmic reticulum: coordination of gene transcriptional and translational controls. Genes Dev 13(10): 1211-33, 1999. Kebache S, Ash J, Annis MG, Hagan J, Huber M, Hassard J, Stewart CL, Whiteway M, Nantel A. Grb10 and active Raf-1 kinase promote Bad- dependent cell survival. J Biol Chem 282:21873-83, 2007. Kedersha N, Anderson P. Stress granules: sites of mRNA triage that regulate mRNA stability and translatability. Biochem Soc Trans 30:963-969, 2002. Kedersha N, Chen S, Gilks N, Li W, Miller IJ, Stahl J, Anderson P. Evidence that ternary complex (eIF2-GTP-tRNA(i)(Met))-deficient preinitiation complexes are core constituents of mammalian stress granules. Mol Biol Cell 13: 195- 210, 2002. Kedersha NL, Gupta M, Li W, Miller I, Anderson P. RNA binding proteins TIA-1 and TIAR link the phosphorylation of eIF2-alpha to the assembly of mammalian stress granules. J Cell Biol 147: 1431-1442, 1999. Kelly KJ. Distant effects of experimental renal ischemia/reperfusion injury. J Am Soc Nephrol 14:1549-1558, 2003.

104 Kennedy SG, Kandel ES, Cross TK, Hay N. Akt/protein kinase B inhibits cell death by preventing the release of cytochrome c from mitochondria. Mol Cell Biol 19(8): 5800–5810, 1999. Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer (4):239-57, 1972. Khabar KS. The AU-rich transcriptome: more than interferons and cytokines, and its role in disease. J Interferon Cytokine Res 25:1-10, 2005. Kimball SR: Eukaryotic initiation factor elF2. Int J Biochem Cell Biol 3125-29, 1999. Kimball SR, Horetsky RL, Ron D, Jefferson LS, and Harding HP: Mammalian stress granules represent sites of accumulation of stalled translation initiation complexes. Am J Physiol Cell Physiol 284:273-284, 2003. Kullmann M, Gopfert U, Siewe B, Hengst L. ELAV/Hu proteins inhibit p27 translation via an IRES element in the p27 5’UTR. Genes Dev 16: 3087– 3099, 2002. Kumar R, Azam S, Sullivan JM, Owen C, Cavaener D, Zhang P, Ron D, Harding HP, Chen JJ, Han A, White BC, Krause GS, DeGarcia DJ. Brain ischemia and reperfusion activates the eukaryotic initiation factor 2alpha kinase, PERK. J Neurochem 77(5): 1418-21, 2001. Kuwana H, Terada Y, Kobayashi T, Okado T, Penninger JM, Irie-Sasaki J, Sasaki T, Sasaki S. The phosphoinositide-3 kinase gamma-Akt pathway mediates renal tubular injury in cisplatin nephrotoxicity. Kidney Int 73:430- 45, 2008. Kwon DS, Kwon CH, Kim JH, Woo JS, Jung JS, Kim YK. Signal transduction of MEK/ERK and PI3K/Akt activation by hypoxia/reoxygenation in renal epithelial cells. Eur J Cell Biol 85(11): 1189-99, 2006. Lal A, Kawai T, Yang X, Mazan-Mamczarz K, Gorospe M. Antiapoptotic function of RNA-binding protein HuR effected through prothymosin alpha. EMBO J 24: 1852–1862, 2005. Landry JR, Mager DL, Wilhelm, BT. Complex controls: the role of alternative promoters in mammalian genomes. Trends Genet 19, 640–648, 2003. Leist M, Jaattela M. Four deaths and a funeral: from caspases to alternative mechanisms. Nat Rev Mol Cell Biol 2(8): 589-98, 2001. Levy NS, Chung S, Furneaux H, Levy AP. Hypoxic stabilization of vascular endothelial growth factor mRNA by the RNA-binding protein HuR. J Biol Chem 273: 6417–6423, 1998. Lim MA, Riedel H, Liu F. Grb10: more than a simple adaptor protein. Front Biosci 9:387-403, 2004.

105 Liu F, Roth RJ. Grb-IR: a SH2-domain-containing protein that binds to the and inhibits its function. Proc Natl Acad Sci USA 92:10287-91, 1995. Lopez de Silanes I, Lal A, Gorospe M. HuR: post-transcriptional paths to malignancy. RNA Biol 2: 11–13, 2005. Lopez de Silanes I, Zhan M, Lal A, Yang X, Gorospe M. Identification of a target RNA motif for RNA binding protein HuR. Proc Nat Acad Sci USA 101:2987- 2992, 2004. Loverre A, Ditonno P, Crovace A, Gesualdo L, Ranieri E, Pontrelli P, Stallone G, Infante B, Schena A, Di Paolo S, Capobianco C, Ursi M, Palazzo S, Battaglia M, Selvaggi FP, Schena FP, Grandaliano G. Ischemia-reperfusion induces glomerular and tubular activation of proinflammatory and antiapoptotic pathways: differential modulation by rapamycin. J Am Soc Nephrol 15:2675-86, 2004. Luo G, Hofmann C, Bronckers AL, Sohocki M, Bradley A. Karsenty G. BMP-7 is an inducer of nephrogenesis, and is also required for eye development and skeletal patterning. Genes Dev 9: 2808–2820, 1995. Macías-Silva M, Hoodless PA, Tang SJ, Buchwald M, Wrana JL. Specific activation of Smad1 signaling pathways by the BMP7 type I receptor, ALK2. J Biol Chem 273: 25628–25636, 1998. Martelli AM, Faenza I, Billi AM, Manzoli L, Evangelisti C, Fala F, Cocco L. Intranuclear 3'-phosphoinositide metabolism and Akt signaling: new mechanisms for tumorigenesis and protection against apoptosis? Cell Signal 18:1101-7, 2006. Maxwell P. HIF-1: an oxygen response system with special relevance to the kidney. J Am Soc Nephrol 14: 2712–2722, 2003. Maxwell SR, Lip GY. Reperfusion injury: a review of the pathophysiology, clinical manifestations and therapeutic options. Int J Cardiol 58:95–117, 1997. Mazan-Mamczarz K, Hagner PR, Corl S, Srikantan S, Wood WH, Becker KG, Gorospe M, Keene JD, Levenson AS, Gartenhaus RB. Post-transcriptional gene regulation by HuR promotes a more tumorigenic phenotype. Oncogene 27:6151-63, 2008. Mazroui R, Di Marco S, Clair E, Von Roretz C, Tenenbaum SA, Keene JD, Saleh M, Gallouzi IE. Caspase-mediated cleavage of HuR in the cytoplasm contributes to pp32/PHAP-I regulation of apoptosis. J Cell Biol 180(1):113- 27, 2008. Meng Z, King PH, Nabors LB, Jackson NL, Chen CY, Emanuel PD, Blume SW. The ELAV RNA-stability factor HuR binds the 5’-untranslated region of the human IGF-IR transcript and differentially represses cap-dependent and IRES-mediated translation. Nucleic Acids Res 33: 2962–2979, 2005.

106 Meng F, Liu L, Chin PC, D'Mello SR. Akt is a downstream target of NF-kappa B. J Biol Chem 277:29674-80, 2002. Meshorer E, Toiber D, Zurel D, Sahly I, Dori A, Cagnano E, Schreiber L, Grisaru D, Tronche F, and Soreq H. Combinatorial complexity of 5' alternative acetylcholinesterase transcripts and protein products. J Biol Chem 279, 29740–29751, 2004. Miyamoto S, Rubio M, Sussman MA. Nuclear and mitochondrial signalling Akts in cardiomyocytes. Cardiovasc Res 82:272-85, 2009. Montie HL, Kayali F, Haezebrouck AJ, Rossi NF, Degarcia DJ. Renal ischemia and reperfusion activates the eIF 2 alpha kinase PERK. Biochim Biophys Acta 1741(3): 314-24, 2005. Morrione A, Valentinis B, Li S, Ooi B. Margolis, Baserga R. Grb10: A new substrate of the insulin-like growth factor I receptor. Cancer Res 56:3165-7, 1996. Moutoussamy S, Renaudie F, Lago F, Kelly PA, Finidori J. Grb10 identified as a potential regulator of growth hormone (GH) signaling by cloning of GH receptor target proteins. J Biol Chem 273:15906-12, 1998. Mrena J, Wiksten JP, Thiel A, Kokkola A, Pohjola L, Lundin J, Nordling S, Ristimaki A, Haglund C. Cyclooxygenase-2 is an independent prognostic factor in gastric cancer and its expression is regulated by the messenger RNA stability factor HuR. Clin Cancer Res 11: 7362–7368, 2005. Nabors LB, Gillespie GY, Harkins L, King PH. HuR, a RNA stability factor, is expressed in malignant brain tumors and binds to adenine- and uridine-rich elements within the 3' untranslated regions of cytokine and angiogenic factor mRNAs. Cancer Res. 61:2154-61, 2001. Novoa I, Zhang Y, Zeng H, Jungreis R, Harding HP, Ron D. Stress-induced gene expression requires programmed recovery from translational repression. EMBO J 22(5):1180-7, 2003. Orike N, Middleton G, Borthwick E, Buchman V, Cowen T, Davies AM. Role of PI 3-kinase, Akt and Bcl-2–related proteins in sustaining the survival of neurotrophic factor–independent adult sympathetic neurons. J Cell Biol 154: 995, 2001. Padanilam BJ, Lewington AJ. Molecular mechanisms of cell death and regeneration in acute ischemic renal injury. Curr Opin Nephrol Hypertens 8:15- 19,1999. Pandey A, Duan H, Di Fiore PP, Dixit VM. The Ret receptor protein tyrosine kinase associates with the SH2-containing adapter protein Grb10. J Biol Chem 270:21461-3, 1995. Park KM, Kramers C, Vayssier-Taussat M, Chen A, Bonventre JV. Prevention of kidney ischemia/reperfusion-induced functional injury, MAPK and MAPK

107 kinase activation, and inflammation by remote transient ureteral obstruction. J Biol Chem 277: 2040-2049, 2002. Patel NS, Cuzzocrea S, Collino M, Chaterjee PK, Mazzon E, Britti D, Yaqoob MM, Thiemermann C. The role of cyclooxygenase-2 in the rodent kidney following ischaemia/reperfusion injury in vivo. Eur J Pharmacol 562: 148– 154, 2007. Peng SS, Chen CY, Xu N, Shyu AB. RNA stabilization by the AU-rich element binding protein, HuR, an ELAV protein. EMBO J 17:3461-3470, 1998. Pestova TV, Kolupaeva VG, Lomakin IB, Pilipenkp EV, Shatsky IN, Agol VI, Hellen CU. Molecular mechanisms of translation initiation in eukaryotes. Proc Natl Acad Sci USA 98(13):7029-36, 2001. Ranji AK, Kvaratskhelia M, Boris-Lawrie KA. Biophysical characterization of features of RNA helicase A that confer translational control of retroviral and selected cellular mRNAs. FASEB J 24: 499, 2010. Riedel H. Grb10 exceeding the boundaries of a common signaling adapter. Front Biosci 9:603-18, 2004. Roberts TM, Boris-Lawrie K. The 5' RNA terminus of spleen necrosis virus stimulates translation of nonviral mRNA. J Virol 74:8111-8118, 2000. Roretz CV, Gallouzi IE. PKR/FADD/caspase-8 pathway mediates the pro- apoptotic activity of the RNA binding protein human antigen R (HuR). J Biol Chem 285(22):16806-13, 2010. Russell RA, Y Zeng, Erlwein O, Cullen BR, McClure MO. The R region found in the human foamy virus long terminal repeat is critical for both Gag and Pol protein expression. J Virol 75 :6817-6824, 2001. Ryan MJ, Johnson G, Kirk J, Furstenberg SM, Zager RA, Torok-storb B. HK-2: an immortalized proximal tubule epithelial cell line from normal adult human kidney. Kidney Int 45(1):48-57, 1994. Saleh, A., Makrigiannis, A. P., Hodge, D. L., and Anderson, S. K. Identification of a novel Ly49 promoter that is active in bone marrow and fetal thymus J. Immunol. 168, 5163–5169, 2002. Saraste A, Pulkki K. Morphologic and biochemical hallmarks of apoptosis. Cardiovasc Res 45(3):528-37, 2000. Satake A, Takaoka M, Nishikawa M. Yuba M, Shibata Y, Okumura K, Kitano K, Tsutsui H, Fujii K, Kobuchi S, Ohkita M, Matsumura Y. Protective effect of 17beta-estradiol on ischemic acute renal failure through the PI3K/Akt/eNOS pathway. Kidney Int 73:308-17, 2008. Schumer M, Colombel MC, Sawczuk IS, Gobé G, Connor J, O'Toole K M, Olsson CA, Wise GJ, Buttyan R. Morphologic, biochemical, and molecular evidence of apoptosis during the reperfusion phase after brief periods of renal ischemia. Am J Pathol 140(4): 831–838, 1992.

108 Schwarz C, Hauser P, Steininger R, Regele H, Heinze G, Mayer G, Oberbauer R. Failure of Bcl-2 up-regulation in proximal tubular epithelial cells of donor kidney biopsy specimens is associated with apoptosis and delayed graft function. Lab Invest 82: 941–948, 2002. Sharples, EJ, Patel N, Brown P, Stewart K, Mota-Philipe H, Sheaff M, Kieswich J, Allen D, Harwood S, Raftery M, Thiemermann C, Yaqoob MM. Erythropoietin protects the kidney against the injury and dysfunction caused by ischemia-reperfusion. J Am Soc Nephrol 15:2115-24, 2004. Smoyer WE, Ransom R, Harris RC, Welsh MJ, Lutsch G, Benndorf R. Ischemic acute renal failure induces differential expression of small heat shock proteins. J Am Soc Nephrol 11: 211–221, 2000. Song G, Ouyang G, Bao S. The activation of Akt/PKB signaling pathway and cell survival. J Cell Mol Med 9 (1) 59–71, 2005. Supavekin S, Zhang W, Kucherlapati R, Kaskel FJ, Moore LC, Devarajan P. Differential gene expression following early renal ischemia-reperfusion. Kidney Int 63 :1714–1724, 2003. Suzuki C, Isaka Y, Shimizu S, Tsujimoto Y, Takabatake Y, Ito T, Takahara S, Imai E. Bcl-2 protects tubular epithelial cells from ischemia reperfusion injury by inhibiting apoptosis. Cell Transplant 17: 223–229, 2008. Sweeney R, Fan Q, Yao MC. Antisense ribosomes: rRNA as a vehicle for antisense RNAs. Proc Natl Acad Sci USA 93:8518-8523, 1996. Tettweilera G, Lasko P. A new model for translational regulation of specific mRNAs. Trends Biochem Sci 31(11): 607-10, 2006. Ting CC, Ching CT, Yi TC, Kuen SS, Kuen LM. Adenovirus-mediated bcl-2 gene transfer inhibits renal ischemia/reperfusion induced tubular oxidative stress and apoptosis. Am J Transplant 5(6):1194–1203, 2005. Tsuritani K, Irie T, Yamashita R, Sakakibara Y, Wakaguri H, Kanai A, Mizushima- Sugano J, Sugano S, Nakai K, Suzuki Y. Distinct class of putative “non- conserved” promoters in humans: Comparative studies of alternative promoters of human and mouse genes. Genome Res 17, 1005–1014, 2007. Van der Velden AW, Thomas AA. The role of the 5' untranslated region of an mRNA in translation regulation during development. Int J Biochem Cell Biol 31:87-106, 1999. Van Why SK, Mann AS, Ardito T, Thulin G, Ferris S, Macleod MA, Kashgarian M, Siegel NJ. Hsp27 associates with actin and limits injury in energy depleted renal epithelia. J Am Soc Nephrol 14:98-106, 2003. Wang R, Brattain MG. AKT can be activated in the nucleus. Cell Signal 18:1722- 31, 2006.

109 Wang W, Furneaux H, Cheng H, Caldwell MC, Hutter D, Liu Y, Holbrook N, Gorospe M. HuR regulates p21 mRNA stabilization by UV light. Mol Cell Biol 20: 760–769, 2000. Weinberg JM. The cell biology of ischemic renal injury. Kidney Int 39:476-500, 1991. Xie L, Zheng X, Qin J, Chen Z, Jin Y, Ding W. Role of PI3-kinase/Akt signalling pathway in renal function and cell proliferation after renal ischaemia/reperfusion injury in mice. Nephrology 11:207-12, 2006. Yeap, BB, Voon DC, Vivian JP, McCulloch RK, Thomson AM, Giles KM, Czyzyk- Krzeska MF, Furneaux H, Wilce MC, Wilce JA, Leedman PJ. Novel binding of HuR and poly(C)-binding protein to a conserved UC-rich motif within the 3'-untranslated region of the androgen receptor messenger RNA. J Biol Chem 277:27183-92, 2002. Yoshida T, Kurelia M, Beato F, Min H, Ingelfinger JR, Stears RL, Swinford RD, Gullans SR, Tang SS. Monitoring changes in gene expression in renal ischemia-reperfusion in the rat, Kidney Int 61:1646–1654, 2002. Yoshida T, Tang SS, Hsiao LL, Jensen RV, Ingelfinger J.R, Gullans SR. Global analysis of gene expression in renal ischemia-reperfusion in the mouse. Biochem Biophys Res Comm 291:787–794, 2002. Zapp ML, Green MR. Sequence-specific RNA binding by the HIV-1 Rev protein. Nature 342:714-716, 1989. Zeisberg M, Bottiglio C, Kumar N, Maeshima Y, Strutz F, Muller GA, Kalluri R. Bone morphogenetic protein-7 delays podocyte injury due to high glucose. Am J Physiol Renal Physiol 285:1060–1067, 2003. Zeisberg M, Hanai J, Sugimoto H, Mammoto T, Charytan D, Strutz F, Kalluri R. BMP-7 counteracts TGF- 1−induced epithelial-to-mesenchymal transition and reverses chronic renal injury. Nat Med 9: 964–968, 2003. Zhang X, Zou T, Rao JN, Liu L, Xiao L, Wang PY, Cui YH, Gorospe M, Wang JY. Stabilization of XIAP mRNA through the RNA binding protein HuR regulated by cellular polyamines. Nucleic Acids Res 37:7623-37, 2009. Zheng X, Zhang X, Sun H, Feng B, Li M, Chen G, Vladau C, Chen D, Suzuki M, Min L, Liu W, Zhong R, Garcia B, Jevnikar A, Min WP. Protection of renal ischemia injury using combination gene silencing of complement 3 and caspase 3 genes. Transplantation 82: 1781–1786, 2006.

110