age-associated alterations in the f344xbn rat heart and

171
Marshall University Marshall Digital Scholar eses, Dissertations and Capstones 1-1-2011 Age-Associated Alterations in the F344XBN Rat Heart and the Efficacy of Chronic Acetaminophen Ingestion as a erapeutic Approach Sunil K. Kakarla Marshall University, [email protected] Follow this and additional works at: hp://mds.marshall.edu/etd Part of the Circulatory and Respiratory Physiology Commons , and the Genetic Processes Commons is Dissertation is brought to you for free and open access by Marshall Digital Scholar. It has been accepted for inclusion in eses, Dissertations and Capstones by an authorized administrator of Marshall Digital Scholar. For more information, please contact [email protected]. Recommended Citation Kakarla, Sunil K., "Age-Associated Alterations in the F344XBN Rat Heart and the Efficacy of Chronic Acetaminophen Ingestion as a erapeutic Approach" (2011). eses, Dissertations and Capstones. Paper 681.

Upload: others

Post on 30-Nov-2021

3 views

Category:

Documents


0 download

TRANSCRIPT

Marshall UniversityMarshall Digital Scholar

Theses, Dissertations and Capstones

1-1-2011

Age-Associated Alterations in the F344XBN RatHeart and the Efficacy of Chronic AcetaminophenIngestion as a Therapeutic ApproachSunil K. KakarlaMarshall University, [email protected]

Follow this and additional works at: http://mds.marshall.edu/etdPart of the Circulatory and Respiratory Physiology Commons, and the Genetic Processes

Commons

This Dissertation is brought to you for free and open access by Marshall Digital Scholar. It has been accepted for inclusion in Theses, Dissertations andCapstones by an authorized administrator of Marshall Digital Scholar. For more information, please contact [email protected].

Recommended CitationKakarla, Sunil K., "Age-Associated Alterations in the F344XBN Rat Heart and the Efficacy of Chronic Acetaminophen Ingestion as aTherapeutic Approach" (2011). Theses, Dissertations and Capstones. Paper 681.

AGE-ASSOCIATED ALTERATIONS IN THE F344XBN RAT HEART AND THE

EFFICACY OF CHRONIC ACETAMINOPHEN INGESTION AS A

THERAPEUTIC APPROACH

Dissertation submitted to

the Graduate College of

Marshall University

In partial fulfillment of

the requirements for the degree of

Doctor of Philosophy

in Biomedical Sciences

by

Sunil Kumar Kakarla

Approved by

Eric R. Blough, Ph.D., Committee Chairperson

Todd Green, Ph.D.

Elsa I. Mangiarua, Ph.D.

Nalini Santanam, Ph.D.

Robert Harris, Ph.D.

Department of Physiology, Pharmacology and Toxicology

Joan C Edwards School of Medicine

Marshall University, Huntington, WV

May, 2011

ABSTRACT

AGE-ASSOCIATED ALTERATIONS IN THE F344XBN RAT HEART AND THE

EFFICACY OF CHRONIC ACETAMINOPHEN INGESTION AS A

THERAPEUTIC APPROACH

By

Sunil K. Kakarla

Despite advances in therapeutic and diagnostic strategies cardiovascular disease

still remains the leading cause of death in older people. Recent studies suggest that age-

related elevations in oxidative stress are associated with increases in cardiac apoptosis

and compensatory hypertrophy of the remaining cardiomyocytes. Acetaminophen has

been shown to exhibit cardioprotective effects following ischemia-reperfusion injury by

acting as an antioxidant. Here, we investigate the molecular mechanisms underlying

aging associated cardiac remodeling in male F344XBN rats and examine the efficacy of

chronic acetaminophen ingestion as a therapeutic approach.

Aging in male F344XBN rats is characterized by a significant increase in heart

size, heart weight, heart weight to body weight ratio and cardiomyocytes fiber cross

sectional area. Our findings suggest that aging-associated cardiac hypertrophy in

F344XBN rats is predominantly mediated via activation of the extracellular regulated

kinase 1/2 (ERK1/2) and protein kinase B (Akt) signaling pathways resulting in increased

protein translational signaling and ultimately increased protein synthesis.

To determine the efficacy of chronic acetaminophen treatment in preventing age-

related alterations in cardiac structure and function, male F344XBN rats (27-mo; n=6)

were treated with acetaminophen (30mg/kg/day p.o.) for six months and underwent serial

electrocardiography before and after the completion of the study. Compared to 6-month

and 33-month age matched control rats, chronic acetaminophen ingestion diminished

age-related increases in cardiac ROS levels and the number of TUNEL positive nuclei.

These changes were accompanied by improvements in the Bax/Bcl2 ratio, diminished

evidence of caspase-3 activation and increased phosphorylation of Akt, ERK1/2, p70S6K

and GSK-3β. In addition, acetaminophen treatment diminished age-related increases in

the incidence of arrhythmias and these alterations were associated with diminished

fibrosis and changes in cardiac miRNA expression. Taken together, these results suggest

that acetaminophen may attenuate the age associated deterioration in cardiac structure

and function possibly via diminishing age associated elevation in reactive oxygen species

production.

iv

ACKNOWLEDGEMENTS

Though this dissertation is an individual effort, it would have not been possible

without spiritual, psychological, scientific, financial and technical efforts of lot many

people. I would like to express many thanks to one and all who helped me in realizing

this dissertation.

First of all I would like to express my deepest gratitude to my mentor Dr. Eric R.

Blough for his supervision, advice, and guidance from the very early stage of this

research as well as believing in me throughout the work. His infectious enthusiasm and

unlimited zeal have been major driving forces through my graduate career at Marshall

University. Dr. Blough has been very helpful and easily accessible with a single knock on

his office door to discuss at length about my research. Often times, it surprises me that he

used to keep his work aside to discuss about my research and I would never forget warm

support and affection he showed towards me. Dr. Blough has created an indelible

impression on my personal and professional career. I can’t ask you anymore Eric; thanks

a lot for everything and I am very much proud to have a mentor like you.

I would like to thank my other committee members Dr. Nalini Santanam, Dr. Elsa

Mangiarua, Dr. Todd Green and Dr. Robert Harris for their critical thinking on my

research, valuable comments and suggestions on the dissertation.

It is the most apt situation to express my greatest gratitude to my parents Venkata

Subbaiah Kakarla and Masthanamma Kakarla, my brother Suresh Babu Kakarla for

v

everything they gave. Without their help, support, motivation and confidence this

dissertation would never have realized.

I am extraordinarily fortunate to have my wife Venkata Surekha Kakarla and my

wonderful kids Asmitha Chowdary Kakarla and Venkata Subodh Chowdary Kakarla who

are very understandable and never complained when I am late to home while I work on

my research. Their love, affection and motivation at home drove me this much distance in

my life thus so far. I would like to thank my sister-in-law Lakshmi Praveena, my niece

Dhatri Kakarla and my nephew Samhith Kakarla for their love and affection. I would like

to take this opportunity to thank my in-laws Prasada Rao Ravipati and Radha Ravipati

who always stood by me in difficult times.

It is my immense pleasure to thank my esteemed colleagues Dr. Kevin Rice, Dr.

Arun Kumar, Dr. Miaozong Wu, Devashish Desai, Anjaiah Katta, Jacqueline Fannin,

Sriram Prasad Mupparaju, Anil Gutta, Satyanarayana Paturi, Madhukar Kolli, Ravi

Arvapalli, Murali Gadde, Siva Nalabotu, Brent Kidd, Srinivas Thulluri, Sudarsanam

Kundla, Eli Shleiser, Hari Addagarla, Nandini Manne, Geeta Nandyala, Radhakrishna

Para and Sravanthi Bodapati who were very friendly and helpful in many ways in

successful completion of my research work. I would like to extend my thanks to Lucy

Dornon who is very helpful in collecting the data that I used for echocardiography

studies.

I would also like to convey my thanks to family friends and well-wishers Manjula

Mupparaju, Kiran Kodali, Satish Adusumilli, Dhanvanthari Emmadi, Dr. Micheal Dyer

vi

and Dr. Kelly Pinkston who were very helpful in my personal life. Finally, I once again

thank Dr. Blough for the grant support.

vii

DEDICATION

To my loving Brother Suresh Babu Kakarla who extended tremendous support to

materialize my dream of pursuing extraordinary career in the USA; has been greatly

helpful in every shade of both my personal and professional life and he always

encouraged me to achieve the best in my life with his valuable suggestions and

motivational thoughts.

viii

TABLE OF CONTENTS

ABSTRACT ....................................................................................................................... ii

ACKNOWLEDGEMENTS ............................................................................................ iv

DEDICATION................................................................................................................. vii

LIST OF FIGURES ......................................................................................................... xi

ABBREVIATIONS ........................................................................................................ xiv

CHAPTER 1 .......................................................................................................................1

INTRODUCTION......................................................................................................... 1

SPECIFIC AIMS .......................................................................................................... 5

CHAPTER II ......................................................................................................................7

REVIEW OF THE LITERATURE ............................................................................ 7

2.1 Aging..................................................................................................................... 7

2.2 Physiological changes with aging ......................................................................... 9

2.3 Cardiac aging ...................................................................................................... 10

2.3.1 Age associated alterations in cardiac function ................................................. 11

2.3.2 Cardiac aging in animal models ....................................................................... 12

2.3.3 Role of oxidative stress in cardiac aging ......................................................... 14

2.3.3.1 Signaling mechanisms underlying cardiac oxidative-nitrosative stress........ 15

2.3.4 Age-associated cardiac apoptosis .................................................................... 17

2.3.4.1 Signaling mechanisms underlying cardiac apoptosis ................................... 19

2.3.5 Age-associated cardiac hypertrophy ................................................................ 20

2.3.5.1 Signaling mechanisms underlying cardiac hypertrophy ............................... 21

2.3.6 Age-associated myocardial fibrosis ................................................................. 23

2.3.6.1 Signaling mechanisms underlying cardiac fibrosis....................................... 24

2.4 Regulation of miRNA in aged hearts .................................................................. 25

ix

2.5 Acetaminophen ................................................................................................... 27

2.5.1 Mode of Action ................................................................................................ 28

2.5.2 Pharmacology .................................................................................................. 29

2.5.3 Cardioprotective value of acetaminophen ....................................................... 29

CHAPTER III ..................................................................................................................33

PAPER 1 ...........................................................................................................................33

Mechanisms of age-related cardiac hypertrophy in the F344XBN rat model ...........34

Abstract ........................................................................................................................ 35

Introduction ................................................................................................................. 36

Materials and Methods ............................................................................................... 38

Results .......................................................................................................................... 42

Aging in the F344XBN rats is associated with cardiac hypertrophy ........................ 42

Aging associated cardiac hypertrophy in F344XBN rats is associated with increases

in the activation of the Ras/Raf/MEK/ERK signaling cascade ................................ 43

PI3K/Akt signaling in aging associated cardiac hypertrophy in the F344XBN rat .. 43

Age-related cardiac hypertrophy in F344XBN rats is associated with increased

protein translational signaling ................................................................................... 45

Discussion..................................................................................................................... 46

PAPER 2 ...........................................................................................................................61

Chronic acetaminophen attenuates age-associated increases in cardiac ROS and

apoptosis in the Fischer Brown Norway rat ..................................................................62

Abstract ........................................................................................................................ 63

Introduction ................................................................................................................. 64

Materials and Methods ............................................................................................... 66

Data Analysis ............................................................................................................... 70

Results .......................................................................................................................... 71

Acetaminophen decreases indices of cardiac ROS ................................................... 71

x

Acetaminophen decreases age-associated cardiomyocyte apoptosis ........................ 72

Acetaminophen attenuates age-associated alterations in cell survival signaling ...... 73

Discussion..................................................................................................................... 75

PAPER 3 ...........................................................................................................................91

Anti-arrhythmic effect of chronic acetaminophen treatment in the aging F344XBN

rat involves diminished myocardial fibrosis and altered miRNAs regulation ...........92

Abstract ........................................................................................................................ 93

Introduction ................................................................................................................. 94

Materials and Methods ............................................................................................... 97

Data Analysis ............................................................................................................. 101

Results ........................................................................................................................ 102

Aging is associated with increased incidence of cardiac arrhythmias .................... 102

Aging is associated with greater myocardial fibrosis ............................................. 102

Aging is associated with diminished connexin-43 levels ....................................... 103

Aging is associated with alterations in cardiac miRNA expression ....................... 103

Acetaminophen treatment is associated with alterations in miRNA expression .... 104

Discussion................................................................................................................... 105

CHAPTER IV.................................................................................................................119

GENERAL DISCUSSION ............................................................................................119

Age-related alterations in cardiac structure and function of F344XBN rats .......... 120

Cardioprotective properties of acetaminophen in aged F344XBN rats .................. 122

Regulation of miRNA with aging and acetaminophen treatment in F344XBN rats

................................................................................................................................. 126

SUMMARY ....................................................................................................................127

FUTURE DIRECTIONS ...............................................................................................128

REFERENCES ...............................................................................................................130

xi

CURRICULUM VITAE ................................................................................................148

LIST OF TABLES

Table 1. Total body weight (BW), heart weight (HW) and heart weight to body weight

ratio (HW/BW %) of 6-, 33-month control and 33-month acetaminophen treated

male F344XBN rats.. ................................................................................................ 80

LIST OF FIGURES

Figure 1. Molecular structure of acetaminophen .............................................................. 28

Figure 2. 1. Heart size, heart weight, body weight, heart weight to body weight ratio

altered with age in F344XBN rats. ........................................................................... 52

Figure 2. 2. Aging in F344XBN rats is associated with cardiomyocyte hypertrophy. ..... 53

Figure 2. 3. Aging is associated with higher phosphorylation of ERK1/2, ERK5 and total

calcineurin protein in F344XBN rat hearts. .............................................................. 54

Figure 2. 4. Aging is associated with higher phosphorylation of Akt in F344XBN rat

hearts. ........................................................................................................................ 55

Figure 2. 5. Aging is associated with higher phosphorylation of GSK3β and PTEN in

F344XBN rat hearts. ................................................................................................. 56

Figure 2. 6. Aging is associated with higher phosphorylation of mTOR in F344XBN rat

hearts. ........................................................................................................................ 57

xii

Figure 2. 7. Aging lowers the phosphorylation of TSC2 and is associated with higher

expression of Raptor in F344XBN rat hearts. ........................................................... 58

Figure 2. 8. Aging is associated with higher phosphorylation of 4EBP1 and rpS6 in

F344XBN rat hearts. ................................................................................................. 59

Figure 2. 9. Proposed scheme depicting the possible molecular mechanisms underlying

age-associated cardiac hypertrophy in F344XBN rats. ............................................. 60

Figure 3. 1. Effect of aging and acetaminophen treatment on indices of cellular ROS. ... 81

Figure 3. 2. Acetaminophen treatment decreases the incidence of TUNEL positive nuclei

in aged cardiac muscle. ............................................................................................. 82

Figure 3. 3. Acetaminophen treatment alters the ratio of Bax / Bcl-2 in aged cardiac

muscle. ...................................................................................................................... 83

Figure 3. 4. Acetaminophen treatment decreases the age-associated activation of caspase-

3................................................................................................................................. 84

Figure 3. 5. Acetaminophen treatment increases Akt phosphorylation in aged cardiac

muscle. ...................................................................................................................... 85

Figure 3. 6. Acetaminophen treatment increases mTOR phosphorylation in aged cardiac

muscle. ...................................................................................................................... 86

Figure 3. 7. Acetaminophen treatment increases ERK1/2 phosphorylation in aged cardiac

muscle. ...................................................................................................................... 87

Figure 3. 8. Acetaminophen treatment increases p70S6K phosphorylation in aged cardiac

muscle. ...................................................................................................................... 88

Figure 3. 9. Acetaminophen treatment increases GSK-3β phosphorylation in aged cardiac

muscle. ...................................................................................................................... 89

xiii

Figure 3. 10. Proposed scheme for the effects of chronic acetaminophen treatment in

attenuating aging-associated cardic ROS and apoptosis. .......................................... 90

Figure 4. 1. Chronic acetaminophen treatment decreases incidence of cardiac

arrhythmias. ............................................................................................................ 113

Figure 4. 2. Chronic acetaminophen attenuates myocardial fibrosis in aged rat hearts. . 114

Figure 4. 3. Chronic acetaminophen preserves myocardial membrane Cx43. ............... 115

Figure 4. 4. Aging decreases myocardial miR-1 and miR-133 expression. ................... 116

Figure 4. 5. Aging decreases myocardial miR-30a and miR-30d expression. ................ 117

Figure 4. 6. Chronic acetaminophen treatment alters myocardial miR-214 expression. 118

xiv

ABBREVIATIONS

4EBP1 Eukaryotic translation initiation factor 4E-binding protein 1

4-HNE 4-hydroxy-2-nonenal

•O2-

Superoxide anions

•OH Hydroxyl radicals

AAALAC Association for assessment and accreditation of laboratory animal care

ANOVA One-way analysis of variance on ranks

Apaf-1 Apoptotic protease activating factor-1

APAP N-acetyl p-aminophenol

ASK-1 Apoptosis-signaling kinase 1

Bax Bcl-2-associated X protein

Bcl2 B-cell lymphoma 2

BSA Bovine serum albumin

Cx43 Connexin 43

COX3 Cyclooxygenase 3

CTGF Connective tissue growth factor

ECL Enhanced chemiluminescence

xv

ERK Extracellular signal-regulated kinase

F344XBN Fisher 344 Brown Norway hybrid

FAK Focal adhesion kinase

GSK-3β Glycogen synthase kinase 3 beta

JNK Jun-N-terminal kinase

KRB Krebs-Ringers Buffer Solution

MAPK Mitogen-activated protein kinase

mTOR Mammalian target of rapamycin

NFAT Nuclear factor of activated T-cells

NF-κB Nuclear factor kappa-light-chain-enhancer of activated B cells

p70S6K p70S6 kinase

PBS Phosphate buffered saline

PBST Phosphate buffered saline with 0.5% tween

PI3K Phosphatidylinositol 3-kinases

PKB Protein kinase B

PKC Protein kinase C

PTEN Phosphatase and tensin homolog

xvi

ROS Reactive oxygen species

rpS6 Ribosomal protein S6

SDS-PAGE Sodium dodecyl sulfate-polyacrylamide gel electrophoresis

TBS Tris buffered saline

TBST Tris buffered saline with 0.5% tween

TGF-β Transforming growth factor beta

TNF-α Tumor necrosis factor-α

TSC2 Tuberous sclerosis protein 2

TUNEL TdT-mediated dUTP nick end labeling

1

CHAPTER 1

INTRODUCTION

The number of elderly, especially in developed countries, continues to grow at a

rapid pace due to availability of better healthcare facilities and improved standards of

living. Epidemiological studies suggest that one in every four citizens of the United

States will be over the age of 65 by the year 2030. This is thought to represent an increase

of 52 million elderly individuals. Furthermore, the population of people over the age of

85 years is expected to increase by 389%, leaving a tremendous burden on the health care

system, as the incidence of cardiovascular diseases increases linearly with age [1].

Indeed, recent data has suggested that older patients account for up to a quarter of all

emergency department visits and that cardiovascular disease remains one of the frequent

presenting complaints [2]. In the United States, cardiovascular disease, e.g.,

atherosclerosis and hypertension that lead to heart failure and stroke, is the leading cause

of mortality, accounting for over 40 percent of deaths in those aged 65 years and above

[3]. Apart from the medical challenge, this impending demographic shift also poses a

major economical challenge for the nation, as the cost of providing health care for elderly

individuals is three to five times greater than the cost for young people [4]. As such, the

development of newer therapeutic agents and methodologies for preventive care is a top

priority. To achieve this goal, it is imperative that we better understand how aging affects

cardiovascular physiology and further, that we increase our knowledge of the underlying

molecular mechanism(s) involved.

2

As we age, numerous physiological and environmental factors increase the risk of

cardiac damage. One such factor is increases in oxidative stress, which has been shown to

cause alterations in the structure and function of major organs, including the heart. It is

well established that the aged heart has a diminished functional and adaptive reserve

capacity, an increased susceptibility to incur damage (e.g., as a result of ischemia), and a

limited practical ability for repair/regeneration [5]. Aging of the heart is mainly

characterized by an increase in the loss of cardiomyocytes and hypertrophy of the

remaining cardiomyocytes, ultimately leading to diastolic heart failure [1]. Throughout

life, the heart is exposed to minor, highly controlled episodes of ischemia followed by

reperfusion, an innate mechanism that helps condition the heart gradually to prevent

injury from such episodes of larger magnitude taking place later in life. This process is

known as ischemic preconditioning of the myocardium [6]. As we age, the capability of

the myocardial tissue to handle oxidative stress decreases progressively. This finding has

led some to postulate that antioxidants may be useful for the treatment and / or prevention

of age-associated cardiac dysfunction.

Cardiac structure and function are remarkably similar among mammalian species,

and the use of animal models has been extremely helpful in developing treatment

strategies for alleviating heart disease in humans. Extending animal studies beyond young

adult ages to very old ages may provide similar benefits to cardiovascular health of the

growing aged population. Multiple experimental models have been used to assess the

effects of aging on reactive oxygen species and cardiovascular disease (CVD), including

dogs, the Fisher 344 (F344), the Fischer 344/NNiaHSd X Brown Norway/BiNia

3

(F344XBN) aging rat models, and other species [7-12]. A recent review of aging rat

models concluded that the F344XBN rat strain seems to live longer while also exhibiting

“physiological aging” to a greater degree than others [13-15]. Recent investigation using

the F344XBN rat model has demonstrated that aging in these animals is associated with

an increased incidence of cardiac arrhythmias as well as impairment of systolic and

diastolic function [16, 17]. In addition, other data has shown that the aged F344XBN

heart exhibits increased expression of oxidative-nitrosative stress markers [18]. Reactive

oxygen species (ROS) such as superoxide anions (•O2-) and hydroxyl radicals (•OH) have

been implicated in cardiac dysfunction, remodeling and in cardiomyocyte apoptosis [19].

Recent data has also suggested that aging in the F344XBN heart is associated with

increases in cardiac apoptosis and further that this increase in cell death is associated with

elevations in cardiac ROS [20]. Whether strategies designed to diminish ROS levels are

efficacious in improving age-related cardiac function is not well understood.

Acetaminophen (N-acetyl p-aminophenol) is an analgesic, antipyretic agent

introduced into western medicine over 100 years ago. Although its precise mechanisms

of action are not known, acetaminophen has been shown to cause dose-dependent

reductions in the amount of circulating prostaglandins, to inhibit myeloperoxidase

activity and decrease the oxidation of lipoproteins. In addition, this agent also appears to

confer cardioprotection by blocking the effects of hydroxyl radical, peroxynitrite, and

hydrogen peroxide [21]. Until recently, most investigations on acetaminophen have

focused on its analgesic/antipyretic properties and hepatotoxicity, while the effects of the

drug on other mammalian organ systems, including the heart and circulation, have been

4

largely ignored. In the last few years, studies have begun to shed light on the antioxidant

properties of acetaminophen and its potential cardioprotective value. Being a phenolic

compound [22], acetaminophen exhibits antioxidant properties [23] and it has been

shown to protect the heart against ischemia-reperfusion injury [24-27]. Other data has

suggested that acetaminophen exhibits potent anti-oxidant activity and that an acute bolus

of acetaminophen is effective in preventing ROS-associated cardiac damage in the rat and

dog heart [28, 29]. These findings are important as they suggest that this molecule may

also have application for the prevention of myocardial infarction, which is one of the

leading causes of severe morbidity and death in the aged. Moreover, as the use of

acetaminophen in the aged is increasing [21], additional research is needed to investigate

the effects of this agent on the cardiovascular system. Herein, we propose to (i.)

Investigate the molecular mechanism(s) underlying aging-associated alterations in the

structure and function of F344XBN rat hearts and (ii.) To determine the efficacy of

chronic acetaminophen treatment in preventing age-associated cardiac dysfunction in the

F344XBN rat heart.

5

SPECIFIC AIMS

Advancing age is associated with increased risk of cardiovascular disease and a

progressive decrease in cardiac function [30]. Recent investigation using the Fischer 344

x Brown Norway F1 hybrid (F344XBN) rat model has demonstrated cardiovascular

changes similar to that seen in aging humans including an increased incidence of cardiac

arrhythmias as well as impairment of systolic and diastolic function [16, 17]. In addition,

other data has shown that the aged F344XBN heart exhibits increased expression of

oxidative-nitrosative stress markers [18]. Reactive oxygen species (ROS) such as

superoxide anions (•O2-) and hydroxyl radicals (•OH) have been implicated in cardiac

dysfunction, remodeling and in cardiomyocyte apoptosis [19]. Recent data has also

suggested that aging in the F344XBN heart is associated with increases in cardiac

apoptosis and that this finding is correlated with elevated cardiac ROS levels [20]. Aging

has been shown to be associated with compensatory hypertrophy in humans and animals;

however, the underlying molecular mechanisms remain unclear.

Our long-term goal is to improve our understanding of the molecular mechanisms

underlying aging associated alterations in the structure and function of the heart and to

develop therapeutic strategies to prevent or reverse such age associated cardiac

dysfunction. The central hypothesis of this dissertation is that chronic acetaminophen

ingestion will be associated with decreases in cardiac oxidative stress that will, in turn, be

strongly associated with the prevention of cardiomyocyte cell death and dysfunction.

We plan to test our central hypothesis and accomplish the objective of this study

by pursuing the following three specific aims:

6

SPECIFIC AIM I: To investigate the different signalling mechanisms underlying aging

associated cardiac hypertrophy in F344XBN rats.

SPECIFIC AIM II: To determine if chronic acetaminophen ingestion is able to diminish

age-related increases in indices of oxidative and nitrosative stress and the effect of these

changes, if present, on the incidence of age-related increases in cell death and apoptotic

signaling.

SPECIFIC AIM III: To investigate age associated alterations in the cardiac rhythm in

F344XBN rats and to determine if chronic acetaminophen ingestion is able to preserve

cardiac structure and diminish cardiac arrhythmias.

7

CHAPTER II

REVIEW OF THE LITERATURE

The following chapter presents a review of the current literature concerning the

present study. Specifically, the following areas will be addressed: aging, physiological

changes with aging in different organ systems, cardiac aging, age-related alterations in

cardiac structure and function, and the potential role of oxidative stress, apoptosis,

cardiac hypertrophy, myocardial fibrosis and miRNA during cardiac remodeling. Finally,

the chapter will conclude with a review on the effects of acetaminophen on cardiac

structure and function.

2.1 Aging

Aging, or the process of growing old, is defined as the gradual deterioration in the

function of cells (mitotic cells, such as fibroblasts and post-mitotic cells, such as neurons,

cardiomyocytes etc.) and structural components (such as bone and muscle) which

consequently have a direct impact on the functional ability of organs, biological systems

and ultimately the organism as a whole [31]. Epidemiological studies indicate that the

elderly population of the world, and especially those in developed countries, continue to

grow rapidly due to availability of better healthcare facilities and improved standard of

living [32]. Due to a decline in fertility rate and old age mortality, in recent years there

has been a shift in the age distribution of the population toward older ages [3]. For

instance, a recent report from the United Nations, World Population Ageing 2009,

indicates that the proportion of older persons, aged 60 years or older has been rising

steadily, from 8 % in 1950 to 11 % in 2009, and is expected to reach 22 % in 2050 [33].

8

According to the U.S. Department of Health and Human Services, the older population

aged 65 years or above numbered 38.9 million in 2008. This was an increase of 4.5

million or 13.0% since 1998 [33]. Current estimates project a further increase of 31% by

2030. At present, one in every eight individuals is aged 65 or more with an additional life

expectancy of 18.6 years [33]. Interestingly, the current intensive biomedical anti-aging

studies may further extend the healthy and productive period of human life in the future

[34]. While increased life expectancy is a success story for mankind, at the same time it

poses a great challenge for our health care system due to the growing burden of chronic

illnesses, injuries, and disabilities. As the aging population increases, the prevalence of

disability, frailty, and chronic diseases (Alzheimer’s, cancer, cardiovascular

and cerebrovascular diseases, etc.) is expected to increase dramatically [33, 35].

Chronic diseases, which affect older adults disproportionately, contribute to

disability, diminished quality of life, and increased long term health care costs [36]. In

the United States, approximately 80% of all persons aged >65 years have at least one

chronic condition, and 50% have at least two [33, 37]. In 2001, the leading causes of

death in developed countries were primarily cardiovascular diseases and cancer, followed

by respiratory diseases and injuries [35]. The health-care cost per capita for persons

aged >65 years in the United States and other developed countries is three to five times

greater than the cost for persons aged <65 years, and the rapid growth in the number of

older persons, coupled with continued advances in medical technology, is expected to

create upward pressure on health- and long-term-care spending [4]. Three-quarters of the

total health-care expenditures in the United States is spent on treating chronic disease

($1·7 trillion per year) and 76% of Medicare expenditure goes to beneficiaries with more

9

than five chronic conditions. The rising prevalence of these diseases has also resulted in

bigger out-of-pocket expenses for senior citizens. In 2008, on average, older Americans

spent $4,605 towards out-of-pocket health care expenditures, an increase of 57% since

1998 [37].

2.2 Physiological changes with aging

Aging is inevitable, and as life expectancy increases it becomes more important to

understand the physiological mechanisms associated with the normal aging process.

Cross-sectional studies have indicated that the prevalence of high body weight or obesity

increases with age up to 60 years and then tends to decline due to decreases in lean body

mass, while the increase in body circumference seen with advanced age is mostly due to

redistribution of body fat to the abdominal cavity [38]. Age-related decreases in lean

body mass, which is also known as sarcopenia, is characterized by decreased muscle

mass and muscle strength, and increased muscle fatigability [39]. Sarcopenia contributes

significantly to the morbidity, affects quality of life, and increases health care costs in the

elderly [40]. Decrease in whole body protein turnover, mixed muscle protein synthesis,

myosin heavy chain synthesis, and mitochondrial protein synthesis have been implicated

in age-related sarcopenia [40]. Such age-related decreases in active cellular mass, i.e., fat

free mass or lean body mass, and a concomitant increase in body visceral fat lead to

decreased body metabolic rate [41]. The free radical theory of aging proposed by Harman

has suggested a possible mechanistic link between metabolic rate and aging [31]. He

proposed that normal oxygen consumption inevitably results in the production of oxygen

free radicals, which, in turn, damage important biological molecules (nucleic acids,

10

proteins, and lipids, among others). This damage, if allowed to proceed unchecked,

oftentimes results in death of the organism [42].

Recently, the free radical theory of aging has evolved into what has been termed

the oxidative-stress theory of aging, which is currently the most widely accepted

mechanistic explanation of aging [43]. Different tissues and organs display different

responses to aging, with more oxidative tissue generally having more age-related

changes. For instance, age-related changes in pulmonary function include but are not

limited to increases in residual volume, decreased elasticity of structures, diminished

compliance, decreased number of airway-supporting structures, increase in closing

volumes and increased risk for atelectasis [44, 45]. The structure, function and

morphology of kidneys also changes markedly with age ultimately leading to decreased

glomerular filtration [46]. Aging has been shown to be associated with structural and

functional changes in the cardiovascular system including the remodeling of the arteries,

heart and the microcirculation [30]. Remodeling of arteries produces a thickening of the

intima-media inducing an increase of the pulse wave and aortic impedance, with greater

resistance to ventricular ejection that in turn induces the remodelling of the left ventricle

and ultimately leading to systolic and diastolic dysfunction [47].

2.3 Cardiac aging

The aged (65+ years of age) now comprise the most rapidly growing segment of

the United States population [33]. Cardiovascular disease is the leading cause of

mortality accounting for over 40 percent of deaths in this population [3, 48]. Apart from

other risk factors like smoking, diabetes, obesity, alcohol, etc., aging by itself represents a

11

major risk factor for cardiovascular disease as it is associated with structural and

functional alterations in the heart [49]. Age-related myocardial structural changes include

loss of myocytes, a progressive increase in myocyte cell volume per nucleus, hypertrophy

of the remaining cardiomyocytes, a reduction in the number of conduction cells and wall

thickening [1]. These structural changes are associated with alterations in cardiac

function including diastolic dysfunction, increased systolic arterial pressure and pulse

pressure, decreased maximal heart rate, maximal cardiac output and maximal VO2 [1]. In

addition, the aged myocardium is prone to inadequate oxygenation as the capillary

density does not increase in parallel with the remodeling myocardium [50].

2.3.1 Age associated alterations in cardiac function

It is established that the aged heart becomes slightly hypertrophic and

hyporesponsive to sympathetic (but not parasympathetic) stimuli, so that the exercise-

induced increases in heart rate and myocardial contractility are blunted in older hearts

[49]. Gerstenblith et al. performed echocardiograms on 105 male participants in the NIA

volunteer Longitudinal Study Program and reported that aging in the normal male is

associated with altered LV diastolic filling, increased aortic root diameter and left

ventricle hypertrophy but little change in contractile ability in the resting state [2]. In

another study, using 2-dimensional echocardiograms, Downes et al. examined the effect

of aging on the pattern of LV diastolic filling in 11 elderly subjects (68 +/- 5 years, mean

+/- standard deviation) and 15 normal young adults (31 +/- 7 years) without coronary

artery disease, systemic hypertension, LV hypertrophy or abnormality of LV systolic

function. Their findings suggested that early diastolic filling is reduced in normal aged

12

subjects, even in the absence of coronary artery disease and systolic dysfunction [51].

Bikkina et al. examined the prognostic implications of asymptomatic ventricular

arrhythmias by performing ambulatory electrocardiographic (ECG) monitoring in 2425

men without clinically evident coronary heart disease, 302 men with coronary heart

disease, 3064 women without disease and 242 women with disease. Their findings

suggested that increased incidence of ventricular arrhythmias is associated with a two

fold increase in the risk for all-cause mortality and myocardial infarction or death due to

coronary heart disease [52]. Haider et al. examined the associations of LV mass and LV

hypertrophy with sudden death risk in a large group of middle-aged and elderly subjects

enrolled in the Framingham Heart Study. Their work demonstrated that LV hypertrophy

is an independent risk factor for sudden death [53]. Morales et al. performed quantitative

echocardiography in seventy-eight subjects (47 men; mean age, 51 years; age range, 23-

87 years) without apparent cardiovascular and systemic disease and reported that

advancing age is associated with changes in myocardial structure that are characterized

by a greater echogenicity and loss in tonal organization, possibly due to increased

collagen content within the fibers [54]. Taken together, these data suggest that cardiac

aging in humans is characterized by alterations in LV diastolic filling, ventricular

dimensions, diastolic function and increased incidence of arrhythmias.

2.3.2 Cardiac aging in animal models

Cardiac structure and function are remarkably similar among mammalian species,

and the use of animal models have provided new approaches to study the effects of aging

on the cardiovascular system [55]. Anversa et al. investigated the effect of age on

13

functional characteristics of the heart in Fischer 344 rats at 4, 12, 20, and 29 months of

age, they found that mean arterial pressure, LV systolic and end-diastolic pressures, its

first derivative (dP/dt), heart rate and stroke volume were comparable in rat groups up to

20 months, while LV end-diastolic pressure was elevated and dP/dt and stroke volume

were decreased during the interval from 20 to 29 months, indicating a significant

impairment of ventricular function with senescence [56, 57]. Boluyt et al. performed an

echocardiographic assessment of age-associated changes in chamber dimensions and

systolic and diastolic properties of the female Fischer 344 (F344) rat hearts. They found

that, compared with young adult 4-month rats, there is a significant decrement in the

resting systolic function of the LV, an increase in isovolumic relaxation time (25.0 +/- 7.6

vs. 17.2 +/- 4.4 ms) and decreases in the tissue Doppler peak E waves at the septal

annulus and at the lateral annulus of the mitral valve in 30-month female F344 rats [58].

In recent years, the Fischer 344/Brown Norway F1 (F344/BNF1) rat has been

recommended for age-related studies by the National Institutes on Aging because this

hybrid rat lives longer and has a lower rate of pathological conditions than other inbred

rat strains [17]. Hacker et al. used echocardiographic and hemodynamic analyses to

determine standard values for LV mass, LV wall thickness, LV chamber diameter, heart

rate, LV fractional shortening, mitral inflow velocity, LV relaxation time, and aortic/LV

pressures in male Fischer 344/ Brown Norway F1 hybrid rats from adulthood to the very

aged (n = 6 per 12-, 18-, 21-, 24-, 27-, 30-, 33-, 36-, and 39-month old group). Their

findings suggested that aged rats had an increased LV mass-to-body weight ratio and

deteriorated systolic function. Histological analysis revealed a gradual increase in fibrosis

14

and a decrease in cardiomyocyte volume density with age [16]. Walker et al. performed

two-dimensional ECHO measurements, two-dimensional guided M-mode, Doppler M-

mode, and other recordings from parasternal long- and short-axis views on male Fischer

344/Brown Norway F1 (F344/BNF1) rats aged 6-, 30-, and 36-month and revealed

persistent cardiac arrhythmias in 72% (13/18) of 36-month rats, 10% (1/10) of 30-month

rats and none in 6-month rats [17]. Taken together these findings suggest that aging in

rodents, like humans, is also associated with increased incidence of cardiac arrhythmias,

decrease in ventricular compliance and impairments in systolic and diastolic function.

2.3.3 Role of oxidative stress in cardiac aging

A growing body of emerging evidence suggests that age-related declines in

cardiac function may be associated with age-related increases in oxidative-nitrosative

stress [59]. Oxidative stress is most commonly defined as an imbalance between the

production and scavenging of reactive oxygen species (ROS) [60]. This imbalance is

associated with measurable increases in ROS. It is well established that elevation of

cardiac ROS levels with age have been shown to influence several important factors

including cardiac hypertrophy, structure and function of blood vessels, inflammation,

apoptosis and alteration of the extracellular matrix profile [61-64]. Indeed, Asano et al.

examined the influence of aging on multiple markers of oxidative-nitrosative stress in the

hearts of 6-, 30- and 36-month F344XBN rats and found that indices of oxidative

(superoxide anion [O2•–], 4-hydroxy-2-nonenal [4-HNE]) and nitrosative (protein

nitrotyrosylation) stress were significantly increased in 36-month hearts and these

findings were highly correlated with increases in LV wall thickness [18]. In a follow-up

15

study, Kakarla et al. showed that aging in F344XBN rats is associated with increased

cardiomyocyte apoptosis suggesting the possible role of oxidative-nitrosative stress [20].

Increases in oxidative stress can also elicit oxidative modification of various cellular

components, such as lipids, proteins, DNA and antioxidant enzymes [65, 66]. Being a

post-mitotic tissue, the myocardium is more vulnerable to oxidative stress as it has lesser

ability to up-regulate antioxidant defenses and / or to repair accumulated oxidative

damage compared to other tissues with greater proliferative capacity such as the liver

[67]. Recent work has demonstrated that mitochondrial ROS generation increases in the

heart with advanced age [68-70]. Additional sources of ROS also include the

peroxisomes, cytochrome P-450, xanthine oxidase and nicotinamide dinucleotide

phosphate (reduced) (NADPH) oxidase, although the quantitative significance of those

pathways in generating ROS and causing cellular senescence remains unclear [71]. Apart

from its role in aging, oxidative-nitrosative stress has also been implicated in chronic

cardiovascular diseases, cardiac injury, cell death, and dysrhythmia [72]. The exact

molecular mechanisms underlying age-related increases in cardiac oxidative-nitrosative

stress and whether pharmacological intervention can be used to counteract such changes

remain to be elucidated.

2.3.3.1 Signaling mechanisms underlying cardiac oxidative-nitrosative stress

Reactive oxygen species (ROS)-dependent modulation of intracellular signaling is

implicated in the development of cardiac structural and functional changes including

cardiomyocyte hypertrophy, interstitial fibrosis, contractile dysfunction and cell death

[73]. Growing evidence suggests that ROS-induced signaling pathways play a key role in

16

the development of cardiac hypertrophy either in response to neurohumoral stimuli or

chronic pressure overload [74]. Experimentally, cardiac hypertrophy has been induced in

cultured cardiomyocytes by angiotensin II, endothelin 1, norepinephrine and tumor

necrosis factor-α in an ROS intracellular signaling dependent manner and can be

inhibited by antioxidants [74]. Angiotensin II (AT II) has been shown to induce cardiac

hypertrophy via a G-protein–linked pathway involving ROS-associated activation of

several downstream signals, including the mitogen activated protein kinases (MAPKs),

while inhibition of ROS by antioxidants blocked ATII-mediated cardiac hypertrophy [75,

76]. Several in vitro and animal studies have also implicated the involvement of protein

kinase C (PKC), p38-MAPK, JNK-MAPK, and ERK1/2-MAPK; PI3K; Akt; several

tyrosine kinases (e.g., src and focal adhesion kinase (FAK)); NF-κB, and calcineurin in

ROS-dependent cardiac hypertrophy [77, 78]. Similarly, the redox-sensitive apoptosis-

signaling kinase 1 (ASK-1) is strongly activated by ROS, which in turn activates p38-

MAPK and JNK-MAPK, while deletion of ASK-1 attenuates p38 and JNK activation and

the cardiac hypertrophic response to ATII [79].

Increasing evidence indicates that ROS can mediate apoptosis by a variety of

mechanisms depending upon the level of ROS produced [80]. For instance, in adult

cardiomyocytes, relatively low levels of H2O2 are associated with the activation of

ERK1/2 MAPK and the stimulation of protein synthesis. Conversely, a higher level of

H2O2, while still activating ERK1/2, also activated the JNK, p38 MAPKs and Akt and

induced apoptosis [80]. Using isolated cardiac cells and different reactive oxygen species

generating systems, researchers have shown that different ROS may use distinct apoptotic

17

pathways [81]. Moreover, the age-related accumulation of ROS-induced biomolecules

are thought to play a major role in the development of cardiovascular disease [82].

However, the signaling mechanisms underlying age-related oxidative stress-induced

cardiac pathologies remain not well understood. Further animal studies involving a wide

range of age groups and the use of different anti-oxidants will no doubt fill the gaps in

understanding of the signaling mechanisms underlying age-related oxidative stress

associated cardiac pathologies.

2.3.4 Age-associated cardiac apoptosis

Apoptosis is a specialized form of programmed cell death that is characterized by

nuclear chromatin condensation and DNA fragmentation. Once death occurs, cellular

debris undergo endocytosis by neighboring cells with no spillage of intracellular content

and no inflammatory response [77]. During the last few years, a growing body of

evidence in both humans and animal models suggests that increases in apoptosis due to

varying stimuli play an important role in the pathological remodeling of the heart [83].

Nevertheless, aging itself has been shown to predispose cardiomyocytes to apoptosis

[84]. Olivetti et al. examined the effects of aging on the human myocardium in 67 hearts

obtained from individuals between the ages of 17-90 years who died from causes other

than cardiovascular disease. Regression analysis of their results demonstrated that the

aging process was characterized by a loss of 38 million and 14 million myocyte nuclei/yr

in the left and right ventricular myocardium, respectively [85]. In another study Melissari

et al. measured cardiac dimensions with respect to body weight, and the number of

myocyte nuclei in the ventricles of 45 males and 22 females (17 to 90 years old), who

18

died from causes independent of cardiovascular disease. These researchers found that

aging was characterized by increases in total heart weight, while LV and inter ventricular

septum weights decreased significantly (r = 0.44; p < 0.001). At the structural level, the

number of myocyte nuclei within the left ventricle LV decreased (r = 0.45; p < 0.001),

whereas myocyte cell volume per nucleus increased (r = 0.30; p >0.05) [86]. Anversa et

al. examined the effect of aging in 3, 10 to 12, and 19 to 21 month old Fischer 344 rat

myocardium and reported that between 3 and 10 to 12 months of age, the mean myocyte

cell volume per nucleus increased by 53 % in the left ventricle while the total number of

myocyte nuclei remained constant [87]. Kajstura et al. determined the effect of aging on

cardiomyocyte cell death in Fischer 344 rats aged 3, 7, 12, 16, and 24 months and

reported that apoptosis is more prevalent than necrosis in the later stages of aging. This

study also showed that apoptosis in the left ventricle increased by more than 200% in 24-

month-old compared to 16-month old rats, with no change in necrotic cell death,

indicating an accelerated rate of apoptosis at a later age [88]. Recently, using the

F344XBN aging rat model, our laboratory has demonstrated that compared to young 6-

month hearts, aged 30-month and very aged 36-month hearts exhibited increased TdT-

mediated dUTP nick end labeling positive nuclei (TUNEL+ nuclei), caspase-3 activation,

caspase-dependent cleavage of alpha-fodrin and diminished phosphorylation of protein

kinase B/Akt (Thr 308) [20]. Taken together, these studies suggest that aging in

mammalian hearts is associated with increases in cardiomyocyte apoptosis and, in an

attempt to compensate the functional loss, the remaining myocytes undergo hypertrophy.

19

2.3.4.1 Signaling mechanisms underlying cardiac apoptosis

The factors regulating cardiac apoptosis are not well understood. Oxidative stress

has been shown to activate mitogen-activated protein kinases (MAPKs), which are

thought to regulate, at least in part, the fate of the cell to go through proliferation,

differentiation, or cell death [89-91]. Different studies have shown that the suppression of

ERK and activation of JNK and p38 MAPKs play a role in apoptosis in different cell

lines [90, 91]. An increased expression of JNK- and p38-MAPKs has been shown to

cause apoptosis in H9c2 cells [92], perfused rat hearts [93], as well as ischemia-

reperfusion rat heart [94]. The role of MAPKs signaling in age-related cardiomyocyte

apoptosis remains largely unknown.

In-vitro and in-vivo experimental models using various apoptotic stimuli have

demonstrated that upregulation of proapoptotic proteins Bax, caspase-3 and p53, as well

as downregulation of antiapoptotic protein Bcl-2, control the apoptotic machinery in the

heart [95-98]. Bax is a key pro-apoptotic regulator that can insert itself into the outer

mitochondrial membrane and promote the release of mitochondrial apoptogenic factors

like cytochrome C [98, 99]. Conversely, Bcl-2 is an anti-apoptotic factor that is thought

to block the membrane insertion of Bax by forming Bax/Bcl-2 heterodimers [100-102].

It is postulated that the ratio of Bax to Bcl-2 plays an important role in regulating cellular

apoptosis with cell death being favored as the balance shifts toward Bax [103]. Cytosolic

cytochrome-c couples with apoptotic protease activating factor-1 (Apaf-1), which leads

to the activation of caspase-9. Activated caspase-9 in turn has been shown to cleave

caspase-3 resulting in caspase-3 activation and proteolytic disassembly of the cell [104,

20

105]. On the other hand, inhibition of the phosphatidylinositol-3-kinase (PI-3 kinase)/Akt

pathway has been shown to induce apoptosis in both in vivo animal models and in vitro

cultured cardiomyocytes [106, 107]. Our laboratory has previously reported that the

increase in cardiac apoptosis observed in aging F344XBN rats is associated with

increased activation of proapoptotic proteins JNK1, JNK3, p38 and Bax, and decreased

expression of anti apoptotic proteins Bcl2, Akt and ERK1/2 [18, 20]. Whether the chronic

administration of antioxidants has an effect on the age-related cardiac remodeling is

currently unknown.

2.3.5 Age-associated cardiac hypertrophy

It is thought that aging in both humans and animal models is associated with

alterations in cardiac structure and function including myocyte dropout and a

compensatory hypertrophy of the remaining myocytes [16, 17, 108]. Although, the

increased heart size is initially a compensatory mechanism, sustained hypertrophy can

ultimately lead to decompensation, arrhythmias and contractile dysfunction, and thus

representing an independent risk factor for heart failure [109]. Gerstenblith et al.

performed echocardiograms on 105 male participants (25-84 years of age) and reported

that even in the absence of arterial hypertension there is a moderate increase in the

thickness of the LV wall, suggesting that aging in the normal male is associated with LV

hypertrophy [2]. In another study, Badano et al. performed echocardiograms on 104

physically active, normal, community-dwelling volunteers (68 men and 36 women),

ranging in age from 18 to 84 years and having no evidence of hypertension or

cardiovascular disease. Their data revealed that LV end-diastolic volume (r = 0.25), wall

21

thickness (r = 0.30) and mass (r = 0.37) increased with age [110]. To determine the age-

related changes in normal human hearts during the first 10 decades of life, Kirtzman et al.

measured heart weights, ventricular wall thicknesses, and valve circumferences in 765

autopsy specimens in the hearts of persons 20 to 99 years old. Their findings suggested

that the ratio of septal to LV free wall thicknesses was significantly elevated after the

seventh decade of life [111]. Similar to humans, echocardiographic studies in F344XBN

rats revealed that intrinsic cardiac aging in these rodent models is also associated with LV

hypertrophy, fibrosis, and diastolic dysfunction [16, 17]. The molecular mechanisms

underlying aging associated cardiac hypertrophy remain to be elucidated.

2.3.5.1 Signaling mechanisms underlying cardiac hypertrophy

The signaling mechanisms underlying cardiac hypertrophy have been widely

studied in models of physiological and pathological cardiac hypertrophy due to exercise,

hypertension, work overload and loss of myocytes following ischemic damage [112].

Characteristics of hypertrophic cardiac myocyte growth include an increase in myocyte

cross-sectional area, increased mRNA content, and protein synthesis [113]. Varying

extracellular stimuli have been shown to induce cardiomyocyte growth including peptide

and amine hormones (Ang II, norepinephrine (NE), endothelin-1, and thrombin),

cytokines (interleukin-1b (IL-1b)), interleukin-6, tumor necrosis factor-α (TNF-α), and

mitogens (fibroblast growth factor and epidermal growth factor) [77]. It is thought that

these agonists bind to cell-surface receptors where they activate multiple signal

transduction cascades that link receptor activation to the regulation of hypertrophic

22

growth, including tyrosine kinases (src, focal adhesion kinase), MAPK (ERK1/2, p38,

JNK, ERK5), PKC, calcineurin, PI3-kinase)/Akt, and NF-kB [114-116].

Several previous studies employing in vivo and in vitro models emphasized that

ERK1/2 MAPK plays a key regulatory role in promoting cardiac hypertrophy through the

Ras/Raf/MEK/ERK signaling pathway [110, 116]. Similar to ERK1/2 MAPK, the

activation of ERK5 in cardiomyocytes has also been shown to increase in response to a

variety of hypertrophic stimuli [117, 118]. Using a transgenic mice model, Sanna et al.

demonstrated that the MEK1–ERK1/2 pathway induces cardiac hypertrophy in vivo, at

least in part, by enhancing the transcriptional activity of nuclear factor of activated T

cells (NFAT) [119]. Calcineurin is a calcium-calmodulin activated serine/threonine-

specific phosphatase that is activated by sustained elevations in intracellular calcium and

is thought to mediate the hypertrophic response through its downstream transcriptional

effector NFAT [120]. Furthermore, chronic stimulation of the PI3K/Akt signaling

pathway in response to pressure overload in the rodent heart and following the incubation

of cultured cardiomyocytes with growth factors has also been shown to induce

myocardial hypertrophy [121-125]. In neonatal rat ventricular myocytes (NRVM),

H2O2 has been shown to induce hypertrophy via activation of src, PKC, and p38,

ERK1/2, and JNK MAP kinases and PI3-kinase signaling proteins, suggesting a

possible link between oxidative stress and cardiac hypertrophy [126-128]. Whether there

is a direct relationship between the age-related oxidative stress and cardiac hypertrophy is

currently unclear. Recently, the mammalian target of rapamycin (mTOR) has emerged as

a potentially important regulator of cardiac hypertrophy by promoting protein

23

translational signaling [129, 130]. Rapamycin, an inhibitor of mTOR activity has been

shown to attenuate cardiac hypertrophy caused by aortic banding induced pressure

overload [131], while it has also been shown to block skeletal muscle hypertrophy [132].

Mounting evidence indicates that mTOR induces hypertrophy by increasing protein

synthesis through the control of a number of downstream signaling proteins that regulate

mRNA translation [133] such as the translational repressor proteins 4E-binding protein-1

(4E-BP1) and the ribosomal protein S6 kinase (rp6k) [123]. Whether age-related

cardiomyocyte hypertrophy involves the same molecular mechanisms that have been

documented with varying hypertrophic stimuli in different models of hypertrophy or

involves different mechanisms remains unclear and warrants further experimentation.

2.3.6 Age-associated myocardial fibrosis

Extensive evidence, derived from both clinical and experimental studies, suggests

that aging in both humans and rodents is characterized by a progressive decrease in the

number of cardiomyocytes and the development of compensatory LV hypertrophy and

fibrosis leading to diastolic dysfunction and heart failure with preserved systolic function

[85, 87, 134, 135]. Previous reports suggest that age-related myocardial fibrosis and

ventricular remodeling leads to increased wall stress, decreased elasticity, impaired early

diastolic filling, and reduced rate of ventricular shortening [136-138]. Excessive fibrosis

can also cause decreases in myocardial compliance and may be linked to the development

of ventricular dysfunction and arrhythmias [139]. Age-related cardiomyocyte loss due to

apoptosis and necrosis leads to an increase in interstitial fibrosis in aged hearts [140].

Myocardial fibrosis is mainly due to progressive accumulation of collagen and other

24

extracellular matrix (ECM) proteins [141]. Eghbali et al. examined the total amount and

structure of myocardial collagen as a function of age in the hearts of male Fischer 344

rats and reported that collagen content in the left ventricle increased from 5.5% of total

protein in young Fischer 344 rats to approximately 12% in senescent animals [142].

Other studies have also shown a significant increase in collagen content in old

normocholesterolemic rabbits [143] and in senescent mice [144].

2.3.6.1 Signaling mechanisms underlying cardiac fibrosis

Although it is well established that cardiac fibrosis plays a key role in

deterioration of cardiac function in advanced age, the underlying molecular mechanisms

remain poorly understood. Recent findings suggest that the renin–angiotensin system and

the generation of reactive oxygen species (ROS) may be involved in age-associated

cardiac fibrosis [140]. Mitochondria isolated from senescent rats showed significant

increases in superoxide radical production [145], while over expression of catalase

targeted to mitochondria diminished cardiac hypertrophy and fibrosis and preserved

diastolic function [145]. These findings indicate an important role for ROS in the

pathogenesis of age-associated fibrotic cardiac remodeling. Siwiki et al. exposed

neonatal and adult rat cardiac fibroblasts in vitro to H2O2 or the superoxide-generating

system xanthine plus xanthine oxidase and demonstrated that oxidative stress regulates

the quantity and quality of extracellular matrix by modulating both collagen synthesis and

metabolism [146]. In another study, Cheng et al. treated cultured neonatal rat cardiac

fibroblasts with angiotensin II (Ang II) to examine the role of ROS mediated signaling in

cardiac fibroblasts and reported that ROS are involved in Ang II-induced proliferation

25

and endothelin-1 (ET-1) gene expression [147]. In a parallel experiment, they also

demonstrated that the combination of AT (I) and ET (A) receptor antagonists plus

antioxidants may be beneficial in preventing the formation of excessive cardiac fibrosis

[147]. Previous studies in both humans and animals have shown ROS-mediated up

regulation of MCP-1/CCL2 in ischemic cardiac fibrosis [36, 37]; however, its

involvement in aging-associated cardiac remodeling needs further investigation [148,

149]. Both ROS and angiotensin II have been shown to activate transforming growth

factor (TGF)-b/Smad2/3 signaling pathways and induce pro-inflammatory mediator

expression leading to cardiac fibrosis. In particular, activation of TGF-/Smad2/3

signaling promotes myofibroblast transdifferentiation and collagen deposition [150-155].

2.4 Regulation of miRNA in aged hearts

MiRNAs (miRNAs) are small ~ 22-nucleotide noncoding RNAs that inhibit

transcription or translation by interacting with the 3′ untranslated region (3′UTR) of

target mRNAs [156]. MiRNAs were first discovered in 1993 while conducting a study of

the gene lin-14 in C. elegans development [157]. Immediately, several researchers began

to shed light on the function of miRNAs and revealed that they play fundamental roles in

diverse biological and pathological processes, including cell proliferation, differentiation,

apoptosis, and carcinogenesis in species ranging from C. elegans and D. melanogaster to

humans [158-161]. Rooji et al. induced cardiac remodelling in mice by transverse aortic

constriction or expression of activated calcineurin and examined the expression of

distinctive miRNAs in diseased cardiac tissue. These researchers reported that >12

miRNAs were up- or down-regulated compared to normal mice [162]. Later, several

26

genetic studies conducted in mice implicated the regulatory role of miRNAs in cardiac

remodeling, growth, conductance and contractility [163-168]. Ikeda et al. measured the

expression of 428 miRNAs in three different types of human heart disease (ischemic

cardiomyopathy, dilated cardiomyopathy, and aortic stenosis). Their data suggest that

expression of many miRNAs is altered in heart disease and that different types of heart

disease are associated with distinct changes in miRNA expression [169]. In this context,

several miRNAs including miR-1, 21, 29, 30, 195, 133, 206 and 208, were shown to play

an important role in the process of cardiac remodeling, putatively by regulating changes

in gene expression that accompany pathological cardiac hypertrophy and contractile

dysfunction [162, 164, 165, 170]. Interestingly, muscle specific miR-1, miR-206 and

miR-133 expression has been shown to be upregulated, downregulated, and unchanged

during cardiac remodeling [164-166]. Furthermore, miR-1 has been implicated in the

etiology of ventricular arrhythmias as it is thought that this miRNA post-transcriptionally

represses the expression of connexin 43 (Cx43) [166]. Similarly, miR-206 has also been

postulated to play a role in cardiac remodeling as miR-206 levels have been shown to be

increased in a rat model of cardiac infarction [171] and in regulating the expression of

Cx43 [166]. Recent data suggest that increased miR-133 resulted in the suppression of

protein synthesis and inhibition of hypertrophic growth in cultured neonatal cardiac

myocytes [164]. MiR-133a has also been shown to act as an antiapoptotic factor by

repressing the translation of caspase-9 [172]. Conversely, miR-133 has also been

implicated in the pathogenesis of cardiac arrhythmias as it has been shown that this

molecule can regulate the ether-a-go-go related gene (ERG), which encodes a key portion

of the cardiac potassium channel [173]. Elevated miR-21 has been linked to increased

27

cardiac fibrosis and increased ERK1/2 MAPK activity [174], and its expression is

relatively low in adult cardiomyocytes compared to neonatal cardiomyocytes [174].

Alternatively, data from other studies suggest that miR-21 has an anti-apoptotic effect

and that it is upregulated during cardiomyocyte hypertrophy [175]. Recently, Duisters et

al. investigated the regulation of connective tissue growth factor (CTGF), and showed

that the expression of miR-133 and miR-30 was inversely related to the amount of CTGF

in rodent models of heart disease and in human patients with pathological LV

hypertrophy [176]. The disparities between results among different studies may likely

reflect differences in disease models, extent of disease, myocardial regions sampled,

assay platforms, normalization methods, sample size, control samples, and statistical

analyses. The factors that regulate the expression of miRNA are not clear; however,

recent studies have suggested that alterations in cellular ROS levels may be involved

[172, 177, 178]. Interestingly, the effect of antioxidants on the expression of miRNA

largely remains unclear and warrants further investigation. Similarly, how aging may

affect the expression of miR-1, miR-206 and miR-133 and their role in age-related

cardiac remodeling and arrhythmias remain unclear.

2.5 Acetaminophen

Acetaminophen (N-acetyl-p-amino-phenol; also known as paracetamol; Figure 1)

is one of the most widely used of all drugs and is well established as a standard

antipyretic and analgesic for mild to moderate pain states [179]. It was first synthesized

at Johns Hopkins University in 1877 and was used in clinical medicine in 1893 by Von

Mehring [180]. Since 1955 acetaminophen has been available over the counter without

28

prescription in the United States [180]. During the 1960s and 1970s, increasing concern

was raised about the toxicity of nonprescription analgesics, but in normal use

paracetamol exhibited a consistent safety profile [22]. In the 1980s, when aspirin was

linked to Reye's syndrome, acetaminophen became the mainstay analgesic and antipyretic

for children with a subsequent reduction in the incidence of Reye’s syndrome [179].

Currently, acetaminophen is a first-line drug of choice for pain management and

antipyresis in a variety of patients, including children, pregnant women, the elderly and

those with osteoarthritis, simple headaches, and noninflammatory musculoskeletal

conditions.

Figure 1. Molecular structure of acetaminophen

2.5.1 Mode of Action

Although acetaminophen has been in use for more than 50 years, its precise

mechanism of action remains unclear. It is thought to inhibit prostaglandin synthesis by

inhibition of cyclooxygenase 3 (COX3) in the central nervous system leading to the

antipyretic and analgesic action. It has little effect on the peripheral cyclooxygenase,

explaining its weak anti-inflammatory properties [181, 182].

29

2.5.2 Pharmacology

Acetaminophen is rapidly absorbed from the GI tract. In the liver, acetaminophen

undergoes glucuronidation or sulfation. Part of the drug is hydroxylated to N-acetyl-

benzoquinone-imine (NABI), which is a highly reactive metabolite [183]. Under normal

circumstances, NABI reacts with the sulfhydryl group on glutathione and is excreted in

urine. At very large doses (~7 times the therapeutic dose), glutathione in the liver is

depleted and large amounts of NABI bind covalently with SH groups on hepatic proteins,

permanently impairing their activity [184, 185]. This decrease in activity, if it persists,

can lead to hepatic necrosis, which may be life threatening [186, 187]. Unconjugated

metabolites can also cause renal tubular necrosis [188].

2.5.3 Cardioprotective value of acetaminophen

Despite decades of wide spread use, little is known about the potential

cytoprotective properties of acetaminophen. Until recently, most acetaminophen

investigations have focused on its analgesic/antipyretic properties and hepatotoxicity

while the effects of this drug on other mammalian organ systems, including the heart and

vasculature, have been largely ignored.

When used to block the synthesis of prostaglandin, acetaminophen caused the

concentration-dependent decrements in the production of prostacyclin [189]. Chou et al.

found that by attenuating the activity of myeloperoxidase, which is an important marker

of heart disease in humans [190], acetaminophen significantly reduced the oxidation of

low-density lipoprotein (LDL) in macrophages [191]. With all these preliminary results,

acetaminophen has begun to be considered as an effective antioxidant because of its

30

ability to protect from membrane lipid oxidation by scavenging peroxyl radicals and

peroxynitrite [192]. Nakamoto et al. examined the effect of acetaminophen on acute

gastric mucosal injury induced by ischemia-reperfusion in rats and showed that

acetaminophen (300 and 500 mg/kg) administered intraperitoneally 90 min before the

ischemia significantly reduced the total area of erosions measured 60 minutes after

reperfusion compared to control animals. The drug also inhibited the increase in lipid

peroxide levels induced by ischemia-reperfusion in gastric tissue and the increase in lipid

peroxidation caused by hydroxyl radicals in vitro. With these results, they concluded that

acetaminophen protects the gastric mucosa by averting the damaging effects of oxygen

radicals [193]. Merrill et al. have shown that acetaminophen protects isolated guinea pig

hearts from the damaging effects of myocardial ischemia and reperfusion, that it

preserves myofibrillar ultrastructure, and was associated with the attenuated production

of both hydroxyl radicals and peroxynitrite [29]. In an another study, Merrill et al.

investigated the effect of acetaminophen on pentobarbital pretreated isolated guinea pig

hearts that underwent ischemia-reperfusion and reported that acetaminophen

(0.35mmol/l) significantly reduces the frequency of pentobarbital induced arrhythmias

[27]. Collectively these findings suggest that under selective conditions (e.g., those

causing release of free radicals and other oxidants) acetaminophen can act as an

antioxidant and is able to prevent ventricular arrhythmias [27, 29]. Merrill et al. has

further investigated the cardioprotective efficacy and antioxidant mechanisms of

acetaminophen in global, low-flow myocardial ischemia and reported that acetaminophen

reduced ischemia-mediated protein oxidation. Furthermore, acetaminophen was able to

reduce ventricular dysfunction caused by exogenously administered hydrogen peroxide

31

indicating that antioxidant mechanisms were responsible for its cardioprotective

properties during postischemia-reperfusion [25]. Golfetti et al. showed that this was true

even if acetaminophen was administered chronically or at the onset of reflow [28].

Furthermore, acetaminophen has shown to attenuate peroxynitrite-mediated activation of

matrix metalloproteinase-2 (MMP-2) and subsequent cleavage of troponin I (TnI) in

ventricular myocytes following myocardial ischemia-reperfusion in isolated guinea pig

hearts, and to preserve contractile function [194-196]. Experiments directed at

investigating the effect of acetaminophen treatment on non-ischemic, hypoxic and

reoxygenated myocardium revealed that acetaminophen treated hearts retained a greater

fraction of mechanical function and preserved myocardial ultrastructure compared to the

vehicle treated hearts [24]. In the dog, acetaminophen treatment has been shown to

reduce infarct size following ischemia-reperfusion [197]. In another study, the same

authors investigated the antiarrhythmic properties of acetaminophen in dogs by inducing

arrhythmias either by regional myocardial ischemia and reperfusion or injecting toxic

levels of ouabain. Their results indicated that acetaminophen has previously unreported

antiarrhythmic properties and that the mechanism appears to involve either protection of

mitochondrial Na+-K

+ pumps or protection of function of the pumps at the sarcolemma

and other locations [198]. Hadzimichalis et al. reported that in guinea pig hearts,

ischemia/reperfusion causes a significant increase in mitochondrial permeability

transition pore (MPTP) opening and mitochondrial cytochrome c release, and that

acetaminophen treatment during injury significantly and completely blocks these effects

leading to a significant decrease in late stage apoptotic myocytes [199]. Taken these

studies together, there is cumulative evidence suggesting that acetaminophen confers

32

functional cardioprotection following cardiac insult, including ischemia-reperfusion,

hypoxia-reoxygenation, exogenous hydrogen peroxide and peroxynitrite administration in

different animal models. The mechanistic pathway(s) underlying acetaminophen

mediated cardioprotection may involve its ability to scavenge hydroxyl radical [25],

hydrogen peroxide [200], peroxynitrate [197], myeloperoxidase [191], cyclooxygenase

[189], and other peroxides [193]. Whether the chronic ingestion of acetaminophen is

efficacious in preventing or reducing age-associated cardiovascular dysfunction has not

been investigated.

33

CHAPTER III

The following chapter includes three different research papers describing in detail the

research experiments conducted to test our hypotheses set forth for the specific aims I, II

and III of this dissertation project.

PAPER 1

The following paper corresponds to the specific aim I

34

Mechanisms of age-related cardiac hypertrophy in the F344XBN rat model

Sunil K. Kakarla1,3

, Sudarsanam Kundla2,3

, Madhukar Kolli1,3

, Anjaiah Katta1, Siva K.

Nalabotu1,3

, Emily Whitt3, Kevin M. Rice

2,3,4 and

Eric R. Blough

1,2,3,4

1 Department of Pharmacology, Physiology and Toxicology, Marshall University,

Joan C. Edwards School of Medicine 2

Department of Biological Sciences, Marshall University 3 Center for Diagnostic Nanosystems, Marshall University

4 Department of Exercise Science, Sport and Recreation, College of Education and

Human Services, Marshall University, Huntington WV

Key words: Aging, Myocardial hypertrophy, protein translational signaling, ERK1/2,

Akt

Running Title: Age-related cardiac hypertrophy is associated with increased

ERK/Akt/mTOR signaling.

35

Abstract

Aims: The aging heart undergoes well characterized changes in cardiac structure and

function though the underlying molecular mechanisms remain obscure. Recent studies

suggest that age-related increases in oxidative stress are associated with increases in

cardiac apoptosis and compensatory hypertrophy of the remaining cardiomyocytes. Here

we examine the possible molecular mechanisms underlying age-related cardiac

hypertrophy in 6, 27, 30, 33 and 36-month F344XBN rat hearts. Methods and results:

Compared to 6-months, the heart to body weight ratio remain unchanged in 27 and 30-

month rats, but was significantly increased by 26% and 37% in 33- and 36-month old

rats, respectively (P < 0.05). Immunohistochemical staining for the sarcolemmal protein

dystrophin and cytoskeletal protein actin revealed robust increases in cardiomyocyte fiber

cross sectional area by 85% and 187% in very aged 33- and 36-month animals (P <

0.05). Increases in cardiomyocyte size was accompanied by the hyperphosphorylation of

several different signaling molecules including ERK1/2, ERK5, Akt (Ser473 and

Thr308), GSK3β, PTEN, mTOR (Ser2448), 4EBP1 (Thr 37/46) and rpS6 (Ser235/236)

(P < 0.05). Conclusion: Taken together, these data suggest that aging in the F344XBN

rat heart is associated with the activation of the ERK1/2 and Akt signaling pathways.

(Word Count: 189)

36

Introduction

Cardiovascular disease remains the leading cause of death in the elderly

population and it is estimated that by 2035, nearly one in four individuals in the United

States will be sixty-five years of age or older [201]. The aged heart undergoes well

characterized structural changes leading to a diminished functional and adaptive reserve

capacity, an increased susceptibility to incur damage and a limited practical ability for

repair / regeneration [5]. Cellular and molecular mechanisms that have been implicated in

age-associated changes in myocardial structure and function in humans have been studied

largely in rodents. The Fischer 344/Brown Norway F1 (F344/BNF1) rat is recommended

for age-related studies by the National Institutes on Aging because this hybrid rat lives

longer and has a lower rate of pathological conditions than inbred rats [202]. According

to the probability of aging curves generated by National Institute on Aging, the aged 30-

month and very aged 36-month rats correspond roughly to humans in their sixth, and

eighth decades of life, respectively [17]. We and others have performed

echocardiographic studies in F344XBN rats and have reported that intrinsic cardiac aging

in these rodent models closely recapitulates age-related cardiac changes in humans,

including left ventricular hypertrophy, fibrosis, and diastolic dysfunction [16, 17].

Previous data from our laboratory has also suggested that aging in F344XBN rat hearts is

associated with increases in oxidative-nitrosative stress and apoptosis [18, 20, 203].

Recent studies in the aged heart have shown that to compensate for the functional loss

due to cardiomyocyte death that the remaining cardiomyocytes undergo compensatory

hypertrophy [87].

37

Although, the increased heart size is initially a compensatory mechanism,

sustained hypertrophy can ultimately lead to decompensation, arrhythmias, contractile

dysfunction and thus represents an independent risk factor for heart failure [109]. It is

well established that increased protein synthesis is a key feature of cardiac hypertrophy

and likely underlies the increased cell and organ size observed under this condition [204].

Previous studies have shown that the extracellular signal-regulated kinase 1/2 (ERK1/2)

plays a central regulatory role in promoting cardiac hypertrophy through

Ras/Raf/MEK/ERK signaling cascade both in vivo and in vitro [205-209] . Other studies

suggested a significant role for protein kinase B or Akt, a serine threonine kinase in

promoting cardiac hypertrophy to varying stimuli [123, 210]. Recently, mammalian

target of rapamycin (mTOR) has emerged as potentially important regulator

of cardiac

hypertrophy by promoting protein translational signaling [129, 130]. The molecular

mechanisms underlying cardiac hypertrophy have been very well investigated in models

of physiological and pathological cardiac hypertrophy due to exercise, hypertension,

work overload and loss of myocytes following ischemic damage [112]. However, the

molecular mechanisms underlying aging associated cardiac hypertrophy are not well

understood. A fundamental understanding of the molecular events underlying age-related

cardiac hypertrophy will provide an additional dimension to studies of aged myocardial

biochemistry and function and may be useful in elucidating mechanisms of excitation-

contraction coupling in the aging heart. Using F344XBN rat model, here we investigated

the molecular signaling profile underlying age-related cardiac hypertrophy and report that

very aged hearts exhibit activation of multiple signaling mechanisms during the

hypertrophic response.

38

Materials and Methods

Animals

Animal care and procedures were conducted in accordance with the Animal Use

Review Board of Marshall University using the criteria outlined by the American

Association of Laboratory Animal Care (AALAC) as proclaimed in the Animal Welfare

Act (PL89-544, PL91-979, and PL94-279). Young (6 months, n = 6), adult (27 months, n

= 6), aged (30-months, n = 6), and very aged (33- and 36 months, n = 6) male F344XBN

rats were obtained from the National Institute on Aging. Rats were housed two per cage

in an AALAC -approved vivarium. Housing conditions consisted of a 12 hour: 12 hour

dark – light cycle with temperature maintained at 22 ± 2°C. Animals were provided food

and water ad libitum. Rats were allowed to acclimate to the housing facilities for at least

two weeks before experimentation began. During this time, the animals were carefully

observed and weighed weekly. None of the animals exhibited signs of failure to thrive,

such as precipitous weight loss, disinterest in the environment, or unexpected gait

alterations.

Tissue collection

Rats were anesthetized with a ketamine-xylazine (4:1) cocktail (50 mg/kg, I/P)

and supplemented as necessary for reflexive response before tissue collections. After

midline laparotomy, the heart was removed and placed in Krebs–Ringer bicarbonate

buffer containing; 118 mM NaCl, 4.7 mM KCl, 2.5 mM CaCl2, 1.2 mM KH2PO4, 1.2

mM MgSO4, 24.2 mM NaHCO3, and 10 mM D-glucose (pH 7.4) equilibrated with 5%

CO2 / 95% O2 and maintained at 37°C. Isolated hearts were quickly massaged to remove

39

any blood from the ventricles, cleaned of connective tissue, weighed, and immediately

snap frozen in liquid nitrogen.

Tissue homogenization and determination of protein concentration

Cardiac muscles were homogenized in a Pierce Tissue Protein Extraction Reagent

(T-PER) (10 mL/g tissue; Rockford, IL, USA) that contained protease inhibitors (P8340,

Sigma-Aldrich, Inc., St. Louis, MO, USA) and phosphatase inhibitors (P5726, Sigma-

Aldrich, Inc., St. Louis, MO, USA). After incubation on ice for 30 min, the homogenate

was collected by centrifuging at 12,000 g for 5 min at 4◦C. The protein concentration of

homogenates was determined via the Bradford method (Fisher Scientific, Rockford, IL,

USA). Homogenate samples were boiled in a Laemmli 2× sample buffer (Sigma-Aldrich,

Inc., St. Louis, MO, USA) for 5 min.

SDS-PAGE and immunoblotting

Fifty micrograms of total protein from each sample was separated on a 10%

PAGEr Gold Precast gel (Lonza, Rockland, ME, USA) and then transferred onto a

nitrocellulose membrane using standard conditions [211]. Membranes were stained with

Ponceau S, and the amount of protein quantified by densitometric analysis to confirm

successful transfer of proteins and equal loading of lanes as described previously [212-

214]. For immunodetection, membranes were blocked for 1 h at room temperature in

blocking buffer (5% non-fat dry milk in TBS-T (20mM Tris-base, 150mM NaCl, 0.05%

Tween-20), pH 7.6), serially washed in TBS-T at room temperature, then probed with

antibodies for the detection of Akt (#9272), phospho-Akt (Ser473) (#9271), phospho-Akt

40

(Thr308) (#9275), mTOR (#2972), phospho-mTOR (Ser2448) (#2971), rpS6 (#2217),

phospho-rpS6 (Ser235/236) (#4858), 4E-BP1 (#9452), phospho 4E-BP1 (Thr37/46)

(#9459), eEF2 (#2332), phospho-eEF2 (Thr56) (#2331), Tuberin/TSC2 (#3612),

phospho-Tuberin/TSC2 (Thr1462) (#3617), PTEN (#9552), phospho-PTEN

(Ser380/Thr382/383) (#9552), GSK-3β (#9315) and phospho- GSK-3β (#9336s) (from

Cell Signaling Technology, Inc., Beverly, MA, USA). Membranes were incubated

overnight at 4◦C in primary antibody buffer (5% BSA in TBS-T, pH 7.6, primary

antibody diluted 1:1000), followed by washing in TBS-T (3X 5 min each), and

incubation with HRP- conjugated secondary antibody (anti-rabbit (#7074) or anti-mouse

(#7076), Cell Signaling Technology, Inc., Danvers, MA, USA) in blocking buffer for 1 h.

After removal of the secondary antibody, membranes were washed (3X 5 min each) in

TBS-T and protein bands visualized on reaction with ECL reagent (Amersham ECL

Western Blotting reagent RPN 2106, GE Healthcare Bio-Sciences Corp., Piscataway, NJ,

USA). Target protein levels were quantified by AlphaEaseFC image analysis software

(Alpha Innotech, San Leandro, CA, USA) and normalized to glyceraldehyde 3-phosphate

dehydrogenase (GAPDH).

Immunohistochemistry

Briefly, sections (8 µm) were washed three times with phosphate-buffered saline

(PBS) containing 0.05% Tween-20 (PBS-T), pH 7.4. After incubation for 30 min in a

blocking solution (5% BSA in PBS-T), sections were incubated with specific antisera

diluted in 3% BSA in PSB-T (antibody dilution of 1:100) for 1 h at 24°C in a humidified

chamber and washed three times with PBS-T prior to incubation with FITC labeled anti-

41

rabbit IgG (1:200) for 30 min at 24°C in a humidified chamber. Experiments performed

in parallel omitting either the primary or the secondary antibody was utilized to confirm

specificity and background immunoreactivity. DAPI was included in the secondary

antibody solution at a concentration of 1.5 µg/ml in order to visualize cell nuclei. After a

final PBS wash and mounting, specimens were visualized by epifluorescence using an

Olympus fluorescence microscope (Melville, NY, USA) fitted with 209 and 409

objectives. Images were recorded digitally using a CCD camera and the integrated optical

density determined using the AlphaEase software package (Alpha Innotech, Santa Clara,

CA, USA).

Data analysis

Results are presented as mean ± SEM. Data were analyzed by using the SigmaStat

3.5 statistical program. A one-way analysis of variance was performed for overall

comparisons with the Student–Newman–Keuls post hoc test used to determine

differences between groups. The level of significance accepted a priori was P ≤ 0.05.

42

Results

Aging in the F344XBN rats is associated with cardiac hypertrophy

Compared to 6-month animals, heart size was visibly larger with age in 27-month

animals and appears to be same in 30-month animals, but then appeared significantly

bigger in 33- and 36-month animals (Figure 2.1A). Compared to 6-month animals, the

body weight was significantly increased until 27-months and then gradually decreased

with age in 30-, 33- and 36-month animals but still significantly higher compared to

young 6-month animals (Figure 2.1B). In a similar fashion, the heart weight was

significantly increased with age in 27-month animals and then tends to decline slightly

yet significantly in 30-month animals, while significantly higher in 33- and 36-month

animals compared to 6-month animals (Figure 2.1B). Whereas, the heart weight to body

weight ratio remained unchanged between 6-, 27- and 30-month animals, while it was

significantly higher in 33- and 36-month animals compared to 6-, 27-, and 30-month

animals (Figure 2.1B) (P < 0.05).

To investigate whether increases in heart mass were associated with larger cardiac

myocytes we next measured the muscle fiber cross sectional area (CSA). Because the

myofibres in different areas of the heart are orientated in different directions we imaged

only those areas of the left ventricle where all the muscle fibers were orientated in a

transverse direction. This task was accomplished by using immunofluorescence after

labeling for dystrophin (FITC-green), actin (rhodamine phalloidin-Red) and DAPI (blue)

on tissue cross sections obtained from 6-, 27-, 30-, 33- and 36-month rat hearts.

Compared to that observed in the 6-month animals, our analysis indicated that the

43

average muscle fiber CSA was 20%, 33%, 85% and 187% higher in the 27-, 30-, 33- and

36-month rat hearts, respectively (Figure 2.2) (P < 0.05).

Aging associated cardiac hypertrophy in F344XBN rats is associated with increases

in the activation of the Ras/Raf/MEK/ERK signaling cascade

Several studies have implied that the extracellular signal-regulated kinase 1/2

(ERK1/2) may play a central regulatory role in promoting cardiac hypertrophy through

the Ras/Raf/MEK/ERK signaling cascade [209]. Herein, our immunoblotting results also

suggest that age-related cardiac hypertrophy in the F344XBN rat is associated with a

significant increase in the phosphorylation of ERK1/2. Compared to that observed in the

hearts obtained from 6-month animals, the phosphorylation of ERK1/2 was 19% and

23% lower in 27- and 30-month hearts before increasing by 50% and 263% in the very

aged 33- and 36- month animals (Figure 2.3A) (P < 0.05). In addition to ERK1/2, the

extracellular signal-regulated kinase ERK5 and the serine/threonine-specific phosphatase

calcineurin (CaN) have also been shown to be activated in cardiomyocytes during the

hypertrophic process [117, 119]. Similar to that observed for ERK1/2, it appears that

ERK5 and CaN exhibit a bimodal signaling response with aging as the expression of

these molecules first decreases with age before returning to levels similar to that seen in

the 6-month adult hearts (Figures 2.3B, 3C) (P < 0.05).

PI3K/Akt signaling in aging associated cardiac hypertrophy in the F344XBN rat

Compared to 6-month animals, phosphorylation of Akt at Ser 473 decreased was

31%, 63%, 56% lower and unchanged in 27-, 30-, 33- and 36-month hearts, respectively

(Figure 2.4) (P < 0.05). Similarly, the phosphorylation of Akt at Thr 308 decreased

44

significantly by 21%, 18%, unchanged in 27-, 30-, and 33-month animals before

becoming increased in the very aged 36-month old animals (Figure 2.4) (P < 0.05).

Consistent with our Akt findings, the phosphorylation of GSK-3β was 17% and 31%

lower in 27- and 30-month old animals, unchanged in 33-month animals before

increasing to a level 26% higher than that seen in the 6-month animals at 36-months.

(Figure 2.5A) (P < 0.05). Compared to 6-month animals, the phosphorylation of PTEN

significantly decreased with age in 27-, 30- and 33-month hearts by 45%, 53% and 30%,

respectively while it remain unchanged in the very aged 36-month animals (Figure 2.5B)

(P < 0.05).

The mammalian target of rapamycin (mTOR) has recently emerged as potentially

important regulator of cardiac hypertrophy [130]. Phosphorylation of mTOR at Ser 2448

significantly decreased with age in 27- and 30-month hearts by 30% and 30%, while it

was unchanged and 73% higher in the hearts obtained from 33- and 36-month old

animals (Figure 2.6) (P < 0.05). Compared to 6-month animals, the amount of Tuberin /

TSC2 decreased significantly with aging by an amount of 22%, 23%, 33% in 27-, 30-,

and 33-month animals and unchanged in the very aged 36-month animals (Figure 2.7A)

(P < 0.05). The amount of phosphorylated Tuberin / TSC2 remain was 14%, 26% and

56% lower in 30-, 33- and 36-month animals compared to 6-month animals (Figure 2.7A)

(P < 0.05). When compared to 6-month animals, the cardiac expression of raptor was

24%, 48%, 33% and 27% lower in 27-, 30-, 33- and 36-month animals (Figure 2.7B) (P

< 0.05).

45

Age-related cardiac hypertrophy in F344XBN rats is associated with increased

protein translational signaling

Increase in phosphorylation of mTOR leads to activation of its downstream

targets S6K1 and 4EBP-1 [215]. Phosphorylated 4EBP1 becomes inactive and binds to

and inactivates eIF4E thereby increasing protein translation [216]. Similar to our findings

for mTOR, the phosphorylation of 4EBP1 was 15%, 33%, 32% lower in 27-, 30- and 33-

month animals, while it remained unchanged in the very aged 36-month animals (Figure

2.8A) (P < 0.05). Upon activation by mTOR, S6K1 in turn phosphorylates ribosomal

protein S6 (rpS6) leading to increased ribosomal biogenesis and ultimately protein

translation [217]. Compared to 6-month animals, the phosphorylation of rpS6 was 24%,

67%, 13% and 93% higher in 27-, 30-, 33- and 36-month animals (Figure 2.8B) (P <

0.05).

46

Discussion

In spite of advances in treatment and early diagnostic approaches, cardiovascular

disease still remains the leading cause of death in the elderly. Studies in both humans and

animal models have indicated that aging is associated with alterations in cardiac structure

and function including myocyte dropout and a compensatory hypertrophy of the

remaining myocytes [16, 17, 108]. Similar to that seen in humans, previous work using

the aging F344XBN rat model, has demonstrated aging in these rats is associated with

increases in cardiac oxidative-nitrosative stress, and apoptosis [18, 20]. Here we examine

the signaling processes that may be associated age-related cardiomyocyte hypertrophy.

Heart size and heart weight gradually increased with age until 27-months,

declined slightly in 30-month animals and then regained in 33- and 36-month animals,

while the body weight increased significantly until 27-months and then tends to decline

gradually in 30-, 33- and 36-month rats (Figure 2.1). Whereas the heart to body weight

ratio remain unchanged up to 30 months before increasing significantly and peaking at

36-months of age (Figure 2.1). These findings suggest that the heart seems to grow in a

parallel fashion with body weight until 27-months of age and then declining at 30-months

of age before undergoing pathological hypertrophy in very aged 33- and 36-months of

animals. The decline in heart weight we observed in 30-month animals compared 27-

month animals might be due to increase in cardiomyocyte apoptosis with age, which we

have reported earlier in F344XBN rats [20]. To investigate whether the increases in heart

weight were due to cardiomyocyte hypertrophy or to increases in fibrosis or some other

alteration, we measured the cardiomyocyte cross sectional area. Our data showed that the

47

cardiomyocyte fiber cross sectional areas tends to increase rapidly from 30 – 36 months

before reaching a maximal diameter at 36 months that is nearly three times larger than

that observed in the 6-month adult animals (Figure 2.2). These data are in agreement with

the heart to body weight ratio findings discussed above indicating that the

cardiomyocytes grow in size in a parallel fashion to body weight until 30-months of age

and then rapidly growing in size due pathological cardiac hypertrophy in very aged 33-

and 36-month animals.

The factors regulating cardiac hypertrophy are not fully understood however

several reports have demonstrating a role for the Raf/MEK/ERK signaling pathway [123,

204, 208, 209, 218, 219]. The data of the present study support this contention as ERK1/2

was robustly increased in the 33- and 36-month rat hearts (Figure 2.3A). Similar findings

were also observed for ERK5 (Figure 2.3B). Like ERK1/2, the activation of ERK5 in

cardiomyocytes has been shown to increase in response to a variety of hypertrophic

stimuli [117, 118]. Using a transgenic mice model, Sanna et al., demonstrated that

MEK1–ERK1/2 pathway induces cardiac hypertrophy in vivo, at least in part, by

enhancing the transcriptional activity of NFAT [119]. Calcineurin is a calcium-

calmodulin activated serine/threonine-specific phosphatase that is activated by sustained

elevations in intracellular calcium and is thought to mediate the hypertrophic response

through its downstream transcriptional effector nuclear factor of activated T cells

(NFAT) [120]. Similar to our findings for the activation of ERK1/2 and ERK5, we also

observed a decrease in the amount of calcineurin with age in 27- and 30-month animals

and then tend to recover in very aged 33- and 36-month rat hearts (Figure 2.3C).

48

It is well accepted that the chronic stimulation of the PI3K/Akt signaling pathway

participates in the development of myocardial hypertrophy [121-123]. Indeed, PI3K/Akt

signaling has been shown to be activated in response to pressure overload in the rodent

heart and following the incubation of cultured cardiomyocytes with growth factors [124].

PI3K inhibitors have also been shown to block ligand-induced hypertrophy in cultured

cardiomyocytes [220] while the over-expression of a constitutively active Akt (caAkt) in

the heart has been reported to increase heart weight and cell size [221], [222]. Our data

are in agreement with these findings. Specifically, we observed decreased

phosphorylation (activation) of Akt at Ser 473 and Thr 308 with age in 27- and 30-month

animals, while significantly recovered in 33- and 36-month animals (Figure 2.4). The

decrease in the amount of calcineurin and the phosphorylation of Akt we observed in 30-

month animals compared to 6- and 27-month animals might be due increases in

cardiomyocyte apoptosis in 30-month F344XBN rats that we have reported before and

the underlying molecular mechanisms awaits future investigations [20]. The GSK3β is a

downstream substrate of Akt that is inhibited by Akt-mediated phosphorylation. GSK3β

functions to negatively regulate the activity of GATA4, β-catenin, c-Myc and NFAT

[223] which leads to a blunting of the hypertrophy response [224]. In agreement with this

data, we note an increased phosphorylation GSK3β in very aged and hypertrophied 36-

month F344XBN rat hearts (Figure 2.5A). Phosphatase and tensin homolog (PTEN), has

been shown to function as a negative modulator of PI3K/Akt/mTOR pathway that is

inactivated upon phosphorylation [225]. Consistent with our Akt data, the

phosphorylation of PTEN was found to be decreased with age in 27- and 30-months of

age and then recovered significantly in the very aged 33- and 36-month animals (5B).

49

Taken together, our data suggests that the PI3K/Akt pathway may be involved in

regulating age-associated cardiac hypertrophy in F344XBN rats.

Like Akt, a growing body of evidence suggests that mTOR may play a key role in

regulating cell growth and protein synthesis [129, 226]. Rapamycin, an inhibitor of

mTOR activity has been shown to attenuate pressure overload cardiac hypertrophy

induced by aortic banding [131] and also blocked skeletal muscle hypertrophy [132].

Here we demonstrate a large increase in the amount of mTOR phosphorylation in the

very aged 36-month animals (Figure 2.6). To examine the upstream signaling events that

may be associated with this age-associated activation of mTOR, we next investigated the

effect of aging on the regulation of TSC2 and Raptor. The tuberous sclerosis complex

TSC2 inhibits mTOR when unphosphorylated while the phosphorylation of this molecule

is thought to lead to increased mTOR activity [227]. Unexpectedly, we found a gradual

decrease in the phosphorylation of TSC2 with aging (Figure 2.7A). As decreased TSC2

phosphorylation is hypothesized to decrease mTOR activity, it is possible that the aging-

associated activation of mTOR we observed is not due to alterations in TSC2 regulation.

In addition, why aging-related increases in the phosphorylation of Akt are not associated

with higher levels of TSC2 phosphorylation is currently unclear. Given that recent studies

have suggested a cross talk between Ras/ERK signaling and mTOR during cardiac

hypertrophy [228, 229], the increased phosphorylation of mTOR we see with aging may

be ERK1/2-dependent or due to some other factor. Besides TSC2, it is thought that

Raptor may also function to influence mTOR activity [230]. Unlike our findings for

TSC2, the amount of Raptor was found to decrease with age until 30-months of age and

50

then tend to recover significantly in the very aged 33- and 36-month animals (Figure

2.7B). Whether this increase in Raptor is directly linked to the increased phosphorylation

of mTOR is currently unclear. Future studies directed to investigate the link will no doubt

provide clues for better understanding of the molecular mechanisms that underlie age-

related cardiac hypertrophy.

It is thought that mTOR may be involved in controlling a number of downstream

signaling proteins that regulate mRNA translation [133] such as the translational

repressor proteins 4E-binding protein-1 (4E-BP1) and the ribosomal protein S6 kinase

(rp6k) [123]. Consistent with our findings of an increased mTOR phosphorylation, here

we found that the phosphorylation of 4EBP1 decreased significantly with age until 33-

months before recovering to the level of 6-month animals in very aged 36-month animals

(Figure 2.8A). The phosphorylation of rpS6 was increased significantly in the very aged

36-month animals (Figures 2.8B). Taken together, our current and previous findings [17,

18, 20, 203] suggest that aging in the F344XBN rat heart is associated with diminished

cardiac function, elevations in oxidative stress, increased cardiomyocyte apoptosis and

cardiomyocyte hypertrophy.

In summary, findings from this study suggest that aging in F344XBN rats is

associated with cardiac hypertrophy and the underlying molecular mechanisms probably

involve increased activation of the ERK1/2 and Akt signaling pathways (Figure 2.9). As

shown in schematic diagram (Figure 2.9), advanced aging in F344XBN rat hearts is

associated with increased activation of ERK1/2-Akt signaling pathways leading to

increased activation of mTOR associated signaling mechanisms thereby resulting in

51

increased protein synthesis and ultimately cardiac hypertrophy. How these events and

processes are interrelated is still unclear and will require further investigation. Given the

high prevalence of age-associated heart disease worldwide and the ever growing

expansion of the elderly population it is anticipated that a greater understanding of the

molecular mechanisms associated with age-related cardiac hypertrophy could lead to

valuable improvements in the ability to manage heart disease.

Funding

This study was supported by Department of Energy Grant DE-PS02-09ER09-01 to

E.R.B.

52

Figures

Figure 2. 1. Heart size, heart weight, body weight, heart weight to body weight ratio altered with age in F344XBN rats. A. Image series depicting hearts immediately after excision from the rats. B. Heart weight and heart weight to body weight ratio significantly increased in very aged 33- and 36-month animals compared to 6-, 27- and 30-month animals. Data are mean ± SE (n = 6). abc: Groups without the same letter are significantly different (P < 0.05).

53

Figure 2. 2. Aging in F344XBN rats is associated with cardiomyocyte hypertrophy. Aging in F344XBN rats is associated with cardiomyocyte hypertrophy. A. Immunofluorescence labeling of dystrophin (FITC-green), actin staining (rhodamine phalloidin-red) and nuclei (DAPI-blue) in 6-, 27-, 30-, 33- and 36-month F344XBN rat heart cross sections. Bar = 50µm. B. Muscle fiber cross sectional area (µm2/fiber). Number of fibers measured for 6-, 27-, 30-, 33- and 36-month rats was 640, 423, 311, 235 and 190 respectively. Data are mean ± SE (n = 4). abcd: Groups without the same letter are significantly different (P < 0.05).

54

Figure 2. 3. Aging is associated with higher phosphorylation of ERK1/2, ERK5 and total calcineurin protein in F344XBN rat hearts. Protein isolates of hearts excised from 6-, 27-, 30-, 33- and 36-month rats were analyzed by immunoblotting for changes in total and phosphorylated ERK1/2 (A), ERK5 (B) and total protein content of calcineurin (C). Ponceau S staining of the nitrocellulose membrane along with densitometric analysis of protein present was done to verify equivalent protein loading between the lanes (data not shown). Data are mean ± SE (n = 6). abcde: Groups without the same letter are significantly different (P < 0.05).

55

Figure 2. 4. Aging is associated with higher phosphorylation of Akt in F344XBN rat hearts. Protein isolates of hearts excised from 6-, 27-, 30-, 33- and 36-month rats were analyzed by immunoblotting for changes in total and phosphorylated Akt (Ser 473, Thr 308). Ponceau S staining of the nitrocellulose membrane along with densitometric analysis of protein present was done to verify equivalent protein loading between the lanes (data not shown). Data are mean ± SE (n = 6). abcd: Groups without the same letter are significantly different (P < 0.05).

56

Figure 2. 5. Aging is associated with higher phosphorylation of GSK3β and PTEN in F344XBN rat hearts. Protein isolates of hearts excised from 6-, 27-, 30-, 33- and 36-month rats were analyzed by immunoblotting for changes in total and phosphorylated GSK3β (A) and PTEN (B). Ponceau S staining of the nitrocellulose membrane along with densitometric analysis of protein present was done to verify equivalent protein loading between the lanes (data not shown). Data are mean ± SE (n = 6). abcd: Groups without the same letter are significantly different (P < 0.05).

57

Figure 2. 6. Aging is associated with higher phosphorylation of mTOR in F344XBN rat hearts. Protein isolates of hearts excised from 6-, 27-, 30-, 33- and 36-month rats were analyzed by immunoblotting for changes in total and phosphorylated mTOR (Ser 2448). Ponceau S staining of the nitrocellulose membrane along with densitometric analysis of protein present was done to verify equivalent protein loading between the lanes (data not shown). Data are mean ± SE (n = 6). abcd: Groups without the same letter are significantly different (P < 0.05).

58

Figure 2. 7. Aging lowers the phosphorylation of TSC2 and is associated with higher expression of Raptor in F344XBN rat hearts. Protein isolates of hearts excised from 6-, 27-, 30-, 33- and 36-month rats were analyzed by immunoblotting for changes in total and phosphorylated TSC2 (A) and total protein content of Raptor (B). Ponceau S staining of the nitrocellulose membrane along with densitometric analysis of protein present was done to verify equivalent protein loading between the lanes (data not shown). Data are mean ± SE (n = 6). abcde: Groups without the same letter are significantly different (P < 0.05).

59

Figure 2. 8. Aging is associated with higher phosphorylation of 4EBP1 and rpS6 in F344XBN rat hearts. Protein isolates of hearts excised from 6-, 27-, 30-, 33- and 36-month rats were analyzed by immunoblotting for changes in total and phosphorylated 4EBP1 (A) and rpS6 (B). Ponceau S staining of the nitrocellulose membrane along with densitometric analysis of protein present was done to verify equivalent protein loading between the lanes (data not shown). Data are mean ± SE (n = 6). abcde: Groups without the same letter are significantly different (P < 0.05).

60

Figure 2. 9. Proposed scheme depicting the possible molecular mechanisms underlying age-associated cardiac hypertrophy in F344XBN rats. Molecules shown in the blue were experimentally assessed in this study, while the molecules in the other circles are postulated to be involved in inducing cardiac hypertrophy to varying stimuli in the previous studies. Aging associated hypertrophy in F344XBN rats is predominantly mediated via ERK/Akt/mTOR signaling.

61

PAPER 2

The following paper corresponds to the specific aim II

62

Chronic acetaminophen attenuates age-associated increases in cardiac ROS and

apoptosis in the Fischer Brown Norway rat

Sunil Kakarla1, Jacqueline C. Fannin

1, Saba Keshavarzian

2, Anjaiah Katta

1,

Satyanarayana Paturi2, Siva K. Nalabotu

1, Miaozong Wu

2, Kevin M. Rice

2, Kamran

Manzoor5, Ernest M. Walker Jr.

3, and Eric R. Blough

1, 2, 4

1Department of Pharmacology, Physiology and Toxicology,

2Department of Biological

Sciences, 3Department of Pathology,

4 Department of Exercise Physiology, Marshall

University, Huntington, WV. 5Charleston Area Medical Center, Charleston, WV.

Keywords: acetaminophen; aging; reactive oxygen species; apoptosis; caspase-3;

bax/bcl2 ratio

Running title: Efficacy of chronic acetaminophen ingestion in preventing age-associated

cardiac dysfunction.

Basic Res Cardiol. 2010 Jul; 105(4):535-44

63

Abstract

There is a growing need for pharmacological agents to manage cardiovascular disease in

the rapidly growing elderly population. Here we determine if acetaminophen is

efficacious in decreasing age-related increases in cardiac reactive oxygen species (ROS)

and apoptosis in aging Fischer344XBrown Norway rats. Compared to 6-month control

animals, indices of oxidative (superoxide anion [O2•–] and 4-hydroxy-2-nonenal [4-

HNE]) and nitrosative (protein nitrotyrosylation) stress were markedly increased in 33-

month old rat hearts. Thirty three month animals that had been treated with

acetaminophen (30mg/kg/day p.o. for six months) exhibited diminished age-related

increases in cardiac ROS levels and TUNEL positive nuclei and these changes were

accompanied by improvements in the Bax/Bcl2 ratio, diminished evidence of caspase-3

activation and increased phosphorylation of protein kinase B (Akt), ERK1/2, p70S6K and

GSK-3β. Taken together these results suggest that acetaminophen may attenuate the age

associated increases in cardiomyocyte apoptosis, possibly via diminishing age associated

elevation in ROS production.

64

Introduction

Cardiovascular disease (CVD) is responsible for more than 30% of all deaths

worldwide and heart failure is the leading cause of death in individuals over age 65 [231].

Large cross-sectional epidemiological studies have demonstrated that aging, by itself,

confers a greater risk for CVD than do the other major risk factors such as plasma lipid

levels, smoking, diabetes, or sedentary lifestyle [232, 233]. Although not well

understood, it has been hypothesized that the profound impact of age on the risk of the

occurrence, severity, and prognosis of cardiovascular disease is related to age-associated

changes in cardiovascular structure and/or function [234].

Similar to humans, advanced aging in the Fischer 344 X Brown Norway

(F344XBN) rat is characterized by cardiac remodeling and an impairment of cardiac

function [32]. The molecular mechanisms responsible for these changes are presently

unclear; however recent data have suggested that age-associated increases in cardiac

apoptosis could play a role in the pathological remodeling of the heart and heart failure

[235, 236]. It is thought that this increased apoptotic activity may be regulated, at least in

part, by the increased production of reactive oxygen species (ROS) [237]. Whether

pharmacological intervention during aging is able to diminish age-associated increases in

cardiac ROS and cardiac apoptosis has not been fully elucidated.

Acetaminophen ((N-acetyl p-aminophenol) is a non-narcotic analgesic and

antipyretic agent with weak anti-inflammatory activity that has been used therapeutically

for several decades [22]. Recent work has suggested that acetaminophen exhibits potent

anti-oxidant activity and that an acute bolus of acetaminophen is effective in preventing

65

ROS-associated cardiac damage in the rat and dog heart [28, 29]. On the basis of these

reports and previous data demonstrating that chronic acetaminophen (30 mg/kg body

weight / day) ingestion can be safely (e.g., in the absence of heptatoxicity) used to

decrease age-associated increases in skeletal muscle ROS levels [238], we tested whether

acetaminophen treatment could also decrease cardiac ROS levels and the incidence of

age-related apoptosis.

66

Materials and Methods

Animals

All procedures were performed in accordance with the Marshall University

Institutional Animal Care and Use Committee (IACUC) guidelines, using the criteria

outlined by the American Association of Laboratory Animal Care (AALAC). Fischer

344/NNiaHSD x Brown Norway/BiNia (F344XBN) rats aged 6 and 27 months, were

purchased from the National Institute on Aging colony in Harlan. Rats were housed two

per cage in an AAALAC approved vivarium with a 12 hour light-dark cycle and

temperature maintained at 22 ± 2°C, and fed ad libitum. All animals were allowed to

acclimatize for 2 weeks before initiation of any treatment or procedures. All animals were

examined for precipitous weight loss, failure to thrive or unexpected gait alterations and

animals with apparent abnormalities were removed from the study. Periodic weight

measurements were taken throughout the duration of the study.

Acetaminophen treatment and tissue collection

The F344XBN rats (27-month) were given acetaminophen (30 mg/kg body

weight/day, Sigma-Aldrich, Inc., St. Louis, MO, USA) for 6 months in drinking water.

Age-matched F344XBN rats were maintained as controls. Rats were anesthetized with a

ketamine–xylazine (4:1) cocktail (50 mg/kg, I/P) and supplemented as necessary for

reflexive response before tissue collections. After midline laparotomy, the heart was

removed and placed in Krebs-Ringer bicarbonate buffer (KRB) containing; 118 mM

NaCl, 4.7 mM KCl, 2.5 mM CaCl2, 1.2 mM KH2PO4, 1.2 mM MgSO4, 24.2 mM

67

NaHCO3, and 10mM α-D-glucose, (pH 7.4) equilibrated with 5% CO2 / 95% O2 and

maintained at 37°C. Isolated hearts were quickly massaged to remove any blood from the

ventricles, cleaned of connective tissue, weighed, and immediately snap frozen in liquid

nitrogen.

Dihydroethidium staining

Cardiac muscles were serially sectioned (8 μm) using an IEC Microtome cryostat

and the sections were collected on poly-lysine coated slides. The fluorescent superoxide

indicator dihydroethidium [hydroethidine (HE)] was used to evaluate superoxide levels as

outlined elsewhere [239]. Upon oxidation, dihydroethidium is intercalated within DNA

exhibiting a bright fluorescent red [240]. Briefly, frozen tissue sections were washed with

phosphate-buffered saline (PBS) for 5 min and then incubated with 200 µl of 10 μm of

dihydroethidium (Molecular Probes, Eugene, OR, USA) for 1 h at room temperature.

After washing with PBS (3 × 5 min), fluorescence was visualized under Texas red filter

using an Olympus BX51 microscope (Olympus America, Melville, NY, USA) equipped

with Olympus WH 20× widefield eyepieces and an Olympus UPlanF1 40×/0.75 objective

lens.

Immunohistochemistry

Immunostaining for nitrotyrosine (#9691, Cell Signaling Technology, Beverly,

MA, USA) and 4-HNE (#MHN-020P, JaICA, Shizuoka, Japan) was visualized by

immunofluorescence as outlined by the antibody manufacturer. Briefly, sections were

washed three times with phosphate-buffered saline (PBS) containing 0.5% Tween-20

68

(PBS-T), pH 7.4. After incubation for 30 min in a blocking solution (5% BSA), sections

were incubated with specific antisera diluted in 3% BSA in PSB-T (antibody dilution of

1:100) for 1 h at 24°C in a humidified chamber. After washing three times with PBS-T,

sections were incubated with FITC anti-rabbit IgG (1:200) for 30 min at 24°C in a

humidified chamber. Experiments performed in parallel omitting either the primary or

secondary antibody were utilized to confirm specificity and background

immunoreactivity. DAPI was included in the secondary antibody solution at a

concentration of 1.5 μg/ml in order to visualize cell nuclei. After a final PBS wash and

mounting, specimens were visualized by epifluorescence using an Olympus fluorescence

microscope (Melville, NY, USA) fitted with 20x and 40x objectives. Images were

recorded digitally using a CCD camera and the integrated optical density determined

using the AlphaEase software package (Alpha Innotech, Santa Clara, CA).

TUNEL staining

DNA fragmentation after staining for dystrophin was determined by TdT-

mediated dUTP nick end labeling (TUNEL) according to the manufacturer’s

recommendations (In Situ Cell Death Detection Kit, Roche Diagnostics, and Mannheim,

Germany). TUNEL staining was performed on tissue sections (8 μm) obtained from 6

month control (n=4), 33 month control (n=4), and 33 month treated (n=4) hearts, which

were fixed with 4% paraformaldehyde, washed with PBS (pH 7.4), and then

permeabilized with 0.1% sodium citrate and 0.1% Triton-X. Cross-sections from each

muscle were treated with DNase I to induce DNA fragmentation as a positive control.

Heart sections were blocked with 3% BSA and incubated with anti-dystrophin antibody

69

(NCL-DYS2, Novocastra Vector Laboratories, Burlingame, CA) at a dilution of 1:200 to

visualize the cell membrane. Nuclei were counter stained using DAPI (VECTASHIELD

HardSet Mounting Medium, Vector Laboratories, Burlingame, CA). Muscle cross

sections were visualized by epifluorescence using an Olympus fluorescence microscope

(Melville, NY) fitted with 20X and 40X objectives and images were recorded digitally

using a CCD camera (Olympus, Melville, NY). The number of TUNEL positive nuclei

was counted in three randomly selected regions in each slide. Four different animals were

counted from each group.

Immunoblotting analysis

Portions of individual cardiac muscles homogenized in buffer (T-PER, 2 mL/g

tissue; Pierce, Rockford, IL) containing protease (P8340, Sigma-Aldrich, Inc., St. Louis,

MO, USA) and phosphatase inhibitors (P5726, Sigma-Aldrich, Inc., St. Louis, MO,

USA). After incubation on ice for 30 min, the homogenate was collected by centrifuging

at 12,000 g for 5 min at 4◦C. Protein concentration of homogenates was determined via

the Bradford method (Fisher Scientific, Rockford, IL, USA). Homogenate samples were

boiled in a Laemmli 2X sample buffer (Sigma-Aldrich, Inc., St. Louis, MO, USA) for 5

min. Fifty micrograms of total protein for each sample was separated on a 10% PAGEr

Gold Precast gel (Lonza, Rockland, ME, USA) and then transferred to nitrocellulose

membranes. Gels were stained with a RAPID Stain protein stain reagent (G-Biosciences,

St. Louis, MO, USA) to verify transfer efficiency to membrane. Membranes were stained

with Ponceau S and the amount of protein quantified by densitometric analysis to confirm

successful transfer of proteins and equal loading of lanes as detailed somewhere else [20].

70

Membranes were blocked with 5% milk in Tris Buffered Saline (TBS) containing 0.5%

Tween- 20 (TBST) for 1 h and then incubated with primary antibody overnight at 4◦C.

After washing with TBST, the membranes were incubated with the corresponding

secondary antibodies conjugating with horseradish peroxidase (HRP) (anti-rabbit (#7074)

or anti-mouse (#7076), Cell Signaling Technology, Danvers, MA, USA) for 1 h at room

temperature. Protein bands were visualized following reaction with ECL reagent

(Amersham ECL Western Blotting reagent RPN 2106, GE Healthcare Bio-Sciences

Corp., Piscataway, NJ, USA). Target protein levels were quantified by AlphaEaseFC

image analysis software (Alpha Innotech, San Leandro, CA, USA). Primary antibodies

against Caspase-3 (#9662), cleaved Caspase-3 (#9661S), Bax (#2772), Bcl2 (#2870S) ,

Akt (#9272), p-Akt (Thr308#9275S/Ser473#9271L), mTOR (#2972), p-mTOR (Ser2448,

#2971S), p70 S6K (#9202), p-p70 S6K (Thr421/Ser424) (#9204S), ERK1/2 (#9102) and

p-ERK1/2 (#4377S) were purchased from Cell Signaling Technology (Beverly, MA,

USA).

Data Analysis

Results are presented as mean ± SEM. Data were analyzed by using the

SigmaStat 3.5 statistical program. A one-way analysis of variance (ANOVA) was

performed for overall comparisons with the Student-Newman-Keuls post hoc test used to

determine differences between groups. The level of significance accepted a priori was ≤

0.05.

71

Results

Acetaminophen decreases indices of cardiac ROS

Compared to 6-month animals, body weight was significantly increased with age

(Table 1). Acetaminophen treatment significantly attenuated age-associated increases in

heart weight and heart / body weight ratio (n=6; P < 0.05). Compared to 6-month control

and 33-month treated animals, dihydroethidium reactivity was visibly increased in 33-

month control hearts (Figure 3.1). Because superoxide can also react with nitric oxide to

form other reactive oxygen species such as peroxynitrite, we also examined heart sections

for the presence of protein nitrosylation using an antibody that recognizes proteins and

peptides containing nitrated tyrosine residues. With aging, the amount of immunoreactive

nitro-tyrosine signal was increased (Figure 3.1). Similar to our findings with

dihydroethidium, acetaminophen ingestion appeared to visibly decrease the amount of

protein nitration (Figure 3.1). In an effort to further investigate the effect of chronic

acetaminophen ingestion on cardiac ROS, we also examined the presence of proteins

modified by lipid peroxidation using immunohistochemistry and an antibody that

recognizes the lipid peroxidation metabolite 4-hydroxy-2-nonenal (4-HNE) [241].

Compared to 6-months animals, the aged myocardium was characterized by a marked

increase in the amount of 4-HNE immunoreactivity (Figure 3.1). Relative to the aged

controls, acetaminophen treatment was found to visibly decrease 4-HNE levels (Figure

3.1). Taken together, these data suggest that chronic acetaminophen ingestion is

associated with diminished indices of cardiac ROS.

72

Acetaminophen decreases age-associated cardiomyocyte apoptosis

Excessive oxidative stress is a well characterized stimulus for cellular apoptosis.

To examine this relationship with aging and acetaminophen treatment we used TdT-

mediated dUTP nick end labeling (TUNEL) to determine the number of cardiomyocytes

that exhibited DNA fragmentation. Compared to adult animals, heart sections from the

aged control group showed significant increases in the number of TUNEL positive nuclei

(Figure 3.2).

To extend upon our TUNEL findings we next examined the regulation of Bax and

Bcl-2. Compared to 6-month animals, Bcl-2 protein levels were 40% lower (P < 0.05)

with aging (Figure 3.3). Acetaminophen treatment in aged animals increased Bcl-2

protein levels by 68% (P < 0.05). Conversely, the amount of Bax remains unchanged

with aging or treatment. It is thought that the ratio of Bax to Bcl-2 plays an important role

in regulating the progression to apoptosis with cell death favored as the balance shifts

toward Bax [103]. With aging, the ratio of Bax to Bcl2 was increased by 68% (P < 0.05).

Compared to 33-month control animals, chronic acetaminophen treatment decreased the

Bax to Bcl2 ratio by 35% (P < 0.05) (Figure 3.3).

Caspases are cysteine-dependent aspartate specific proteases that are activated

during cell death [104]. Compared to 6-month control animals, the amount of full length

(inactive) caspase-3 was 22% (P < 0.05) and 36% (P < 0.05) higher in 33-month control

and treated animals (Figure 3.4) (P < 0.05). Caspase-3 activation, as measured by

densitometric analysis of the cleaved 19 and 17 Kda caspase-3 fragments, was similar

between 6-month adult and 33-month old treated animals but 186% and 92% higher (P <

0.05) in the 33-month control hearts (Figure 3.4). Taken together, these data suggest that

73

chronic acetaminophen treatment decreases the incidence of age-related cardiomyocytes

apoptosis.

Acetaminophen attenuates age-associated alterations in cell survival signaling

The Akt/PKB is a serine/threonine protein kinase that functions as a critical

regulator of cell survival through its ability to prevent apoptosis [242]. Acetaminophen

increased Akt Thr308 and Ser473 phosphorylation (activation) by 29% (P<0.05) and

99% (P<0.05), respectively (Figure 3.5). To confirm these findings we also examined the

phosphorylation of the mammalian target of rapamycin (mTOR) on the basis of recent

data suggesting that the phosphorylation of this molecule mediates the activation of Akt

[243]. Compared to untreated control animals, acetaminophen increased mTOR (Ser

2448) phosphorylation by 79% (P<0.05) (Figure 3.6). Recent studies have suggested that

oxidative stress causes decreases in ERK1/2 phosphorylation [244] and that therapeutic

approaches to increase phosphorylation of ERK1/2 can confer cardioprotection [245].

Similar to these studies, six months of acetaminophen treatment significantly increased

phosphorylation of ERK1/2 by 32% compared to that observed in the untreated control

animals (P<0.05) (Figure 3.7). To further confirm this increase in ERK1/2 activity, we

next examined the phosphorylation status of the ERK1/2 substrate, p70S6K, at

Thr421/Ser424 [246]. As expected, acetaminophen treatment significantly increased

p70S6k phosphorylation (Thr421/Ser424) by ~110% compared to that observed in the

age-matched untreated control animals (P<0.05) (Figure 3.8). Like Akt/PKB, glycogen

synthase kinase 3-beta (GSK-3β) has been shown to hinder apoptosis as it can inhibit the

opening of the mitochondrial permeability transition pore (mPTP) when phosphorylated

74

[247]. Compared to untreated control animals, acetaminophen treatment significantly

increased the phosphorylation of GSK-3β (Ser 9) by 42% (P<0.05) (Figure 3.9).

75

Discussion

The age-related accumulation of ROS-induced biomolecules is thought to play a

major role in the development of cardiovascular disease [82]. For instance, both clinical

and experimental evidence have indicated that ROS are increased in chronic heart failure

[248]. Furthermore, antioxidants and free radical scavengers have been shown to

mitigate age-related and other ROS induced cardiac pathologies [249, 250]. In particular,

aspirin is an anti-oxidative compound with clearly demonstrated efficacy in the

prevention of heart disease [251]. Still, prophylactic aspirin therapy fails to fully prevent

the development of heart disease, increases the risk of bleeding problems, and is

associated with other safety concerns [251]. Thus, the prevention of heart disease in the

elderly could be improved significantly by the identification and characterization of

additional compounds that are safe, affordable, and effective for long-term use in

humans. In fact, characterization of the long-term effects of such compounds is crucial in

the development of combinatorial antioxidative treatments, which may eventually

represent the most effective treatment for multiple degenerative diseases [252]. Here, we

demonstrate that the chronic use of acetaminophen acts to diminish the incidence of

cardiomyocyte apoptosis in the aging F344XBN rat model.

Age-related increases in cardiac ROS levels have been implicated in the

pathological remodeling of the heart and heart failure [253]. Previous studies using the

F344XBN rat have demonstrated that aging in these animals is characterized by an

elevation in the amount of cardiac ROS, protein nitration and 4-hydroxy-2-nonenal (4-

HNE) [18]. The findings of the current study support these data while also demonstrating

76

that elevated indices of oxidative stress can be attenuated by chronic acetaminophen

treatment (Figure 3.1). To our knowledge, these findings have not been demonstrated

before.

How acetaminophen might decrease cardiac ROS is not well understood. Previous

reports suggest that acetaminophen may act as an antioxidant as it is a mono-phenol [23].

Possible sources of increased ROS in the aging myocardium have not been fully

elucidated but could include xanthine and NAD(P)H oxidoreductases, cyclooxygenase,

the mitochondrial electron transport chain and activated neutrophils, among many others

[254]. Although many of these sources could potentially produce ROS, it is thought that

increased NADH/NAD(P)H oxidase and / or alterations in mitochondrial function may

be the main cause of augmented ROS generation in the aged heart [255]. Whether age-

related alterations in these systems or others are responsible for the increases in the ROS

indices we see in the aging F344XBN heart is currently being investigated. Similarly,

whether acetaminophen affects these processes or others is not clear. Merrill and

colleagues have shown that acetaminophen significantly reduces ventricular dysfunction

caused by exogenously administered hydrogen peroxide [29] and that it attenuates the

burst of hydroxyl radicals released in the early minutes of reperfusion following global

myocardial ischemia [27]. Chronically administered acetaminophen has also been shown

to provide cardioprotection to the postischemic, reperfused rodent myocardium [28]. In a

recent study using Langendorff-perfused guinea pig hearts, Hadzimichalis and colleagues

demonstrated that administration of acetaminophen just before an ischemic attack can

result in attenuation of functional damage

via inhibition of MPTP opening and

cytochrome c release-induced apoptotic cell death following ischemia/reperfusion [199].

77

Acetaminophen has also been found to be effective in attenuating the deleterious effects

of peroxynitrate [197], myeloperoxidase [191], cyclooxygenase [189], and other

peroxides [193]. Whether acetaminophen targets these or other types of ROS and if this

action, if present, is responsible for the effects we observe in the aging F344XBN heart

will require further study.

Increases in cardiac apoptosis have been implicated in the pathological

remodeling of the heart and heart failure [20]. This increased apoptotic activity may be

mediated by the increased production of ROS [237, 252]. Indeed, oxidative stress seems

to function as a master regulator of apoptosis in cells [252]. In addition to apoptosis,

ROS levels may also act to expand the region of apoptotic activity surrounding damaged

cells during heart attack and stroke [256]. Given the potential linkages between ROS

levels and cardiomyocyte apoptosis we examined the effect of chronic acetaminophen

ingestion on the amount of TUNEL positive cells in heart sections of aged control and

acetaminophen treated animals. Consistent with our findings demonstrating that

acetaminophen can reduce indices of cardiac ROS, we also show that acetaminophen

may be efficacious in reducing the amount of age-related cardiac myocytes apoptosis

(Figure 3.2). In an effort to confirm these findings, we next investigated the ratio of Bax

to Bcl-2. Bax is a key pro-apoptotic regulator that can insert itself into the outer

mitochondrial membrane and promote the release of mitochondrial apoptogenic factors

[99]. Conversely, Bcl-2 is an anti-apoptotic factor that is thought to block the membrane

insertion of Bax by forming Bax/Bcl-2 heterodimers [100]. It is postulated that the ratio

of Bax to Bcl-2 plays an important role in regulating cellular apoptosis with cell death

being favored as the balance shifts toward Bax [103]. Consistent with our TUNEL data,

78

we found that chronic acetaminophen treatment reduces apoptotic Bax / Bcl-2 signaling

in the aged F344XBN rat heart (Figure 3.3).

It has been postulated that cytosolic cytochrome-c couples with apoptotic protease

activating factor-1 (Apaf-1) which leads to the activation of caspase-9. Activated

caspase-9 in turn has been shown to cleave caspase-3 resulting in caspase-3 activation

and proteolytic disassembly of the cell [104]. Similar to previous work [257], and

consistent with our TUNEL and Bax/Bcl-2 data, we also observed an increase in the level

of cleaved caspase-3 with aging (Figure 3.4). In addition, we also demonstrate that

acetaminophen treatment is associated with decreased caspase-3 activation (Figure 3.4).

Whether this decrease in caspase-3 activation by itself, or acting in concert with other

factors is responsible for the decreases in apoptosis we see with acetaminophen treatment

is currently unclear.

To investigate the possibility that molecules other than caspase-3 might also play

a role in regulating the acetaminophen-associated decreases in cardiac apoptosis

observed, we also examined the effects of acetaminophen on the regulation of protein

kinase B (Akt/PKB). The Akt/PKB is a serine/threonine protein kinase that functions as a

critical regulator of cell survival and proliferation [107, 258]. As expected from our

caspase-3 and Bax/Bcl-2 data, we found an increased expression of phosphorylated

(activated) Akt with acetaminophen treatment (Figure 3.5). As previous studies have

shown that mTOR may function to regulate Akt activity [259, 260], we also examined if

acetaminophen ingestion was associated with alterations in the phosphorylation (Ser

2448) of mTOR. Similar to our findings for Akt, we also observed a robust increase in

the amount of phosphorylated mTOR with chronic acetaminophen ingestion (Figure 3.6).

79

Why acetaminophen treatment may be associated with increases in Akt phosphorylation

is not clear as alterations in ROS have been shown to increase [261] or decrease [262,

263] Akt phosphorylation. Similarly, whether a direct linkage exists between changes in

Akt phosphorylation and the acetaminophen-associated changes in caspase-3 activation

we observe is unknown.

It has been suggested that increased phosphorylation of ERK1/2, is associated

with cell survival [245] and oxidative stress has been shown to be linked to diminished

ERK1/2 phosphorylation [244]. Similarly, other work has demonstrated that ERK1/2 can

protect against ischemia-reperfusion induced oxidative stress [264, 265] and that ERK1/2

is involved in regulating the cardioprotective effects of nitric oxide (NO) following

ischemia-reperfusion [266]. Like ERK1/2, activated (phosphorylated) GSK-3β has also

been shown to exhibit anti- apoptotic activity as it may be involved in regulating the

opening of the mitochondrial permeability transition pore [247]. Consistent with our

findings for Akt, here we demonstrate increased phosphorylation of ERK1/2 and GSK-3β

with acetaminophen treatment (Figures. 3.7, 3.9). Whether these changes in ERK1/2 and

GSK-3β are directly associated with the diminished cardiac myocyte apoptosis we

observe in the acetaminophen treated animals is currently unclear but warrants further

investigation.

In summary, we have demonstrated for the first time that chronic acetaminophen

use attenuates excess ROS and apoptotic Bax/Bcl2 signaling in aged cardiac cells,

thereby helping to prevent caspase-mediated apoptosis. These beneficial effects suggest

that chronic acetaminophen usage may protect the heart by promoting the development of

anti-oxidative capacity (Figure 3.10). Still, little is known about the mechanisms of

80

cardioprotection employed by chronic acetaminophen treatment. Increasing our

understanding of these mechanisms and how they may interact with other drugs and

disease processes could lead to new ways in improving our ability to manage heart

disease.

Table 1. Total body weight (BW), heart weight (HW) and heart weight to body weight

ratio (HW/BW %) of 6-, 33-month control and 33-month acetaminophen treated male

F344XBN rats. Results are expressed as gram (g) or HW/BW ratio (mean ± SD; n=6). (*)

indicates significant difference from adult (6-month) value (P<0.05). (†) indicates

significant difference from age matched 33-month control animals (P<0.05).

81

Figures

Figure 3. 1. Effect of aging and acetaminophen treatment on indices of cellular ROS. A.

Indices of oxidative (superoxide anion [O2•–], 4-hydroxy-2-nonenal [4-HNE]) and

nitrosative (protein nitrotyrosylation) stress in 6-month control, 33-month control, and

33-month treated hearts. Bar=100µm. B. Quantification of cardiac ROS as determined by

intensity of fluorescence. Results are expressed relative to the 6-month value. (*)

indicates significant difference from adult (6-month) value (P<0.05). (†) indicates

significant difference from age matched 33-month control animals (P<0.05). n = 4 hearts

per age group.

82

Figure 3. 2. Acetaminophen treatment decreases the incidence of TUNEL positive nuclei

in aged cardiac muscle. A. Double immunofluorescence labeling of dystrophin (Texas

Red), TUNEL (FITC) and nuclei (DAPI) in 6-month control, 33-month control, and 33-

month treated hearts. Bar = 100 μm. B. Number of TUNEL positive nuclei per mm2

in 6-

month control, 33-month control, and 33-month treated hearts. (*) indicates significant

difference from adult (6-month) value (P<0.05). (†) indicates significant difference from

age matched 33-month control animals (P<0.05). n = 4 hearts per age group.

83

Figure 3. 3. Acetaminophen treatment alters the ratio of Bax / Bcl-2 in aged cardiac

muscle. Protein isolates of hearts from 6-month contol, 33-month control and 33-month

treated hearts were analyzed by immunoblotting to determine Bax and Bcl2 protein

levels. Ponceau S staining of the nitrocellulose membrane along with densitometric

analysis of protein present was done to verify equivalent protein loading between the

lanes. (*) indicates significant difference from adult (6-month) value (P<0.05). (†)

indicates significant difference from age matched 33-month control animals (P<0.05). n

= 6 for all groups.

84

Figure 3. 4. Acetaminophen treatment decreases the age-associated activation of caspase-

3. Protein isolates of hearts from 6-month contol, 33-month control and 33-month treated

hearts were analyzed by immunoblotting to determine the amount of full length and

cleaved caspase-3. Ponceau S staining of the nitrocellulose membrane along with

densitometric analysis of protein present was done to verify equivalent protein loading

between the lanes (data not shown). (*) indicates significant difference from adult (6-

month) value (P<0.05). (†) indicates significant difference from age matched 33-month

control animals (P<0.05). n = 6 for all groups.

85

Figure 3. 5. Acetaminophen treatment increases Akt phosphorylation in aged cardiac

muscle. Protein isolates of hearts from 6-month contol, 33-month control and 33-month

treated hearts were analyzed by immunoblotting to determine the amount of total and

phosphorylated Akt (Thr 308, Ser 473). Ponceau S staining of the nitrocellulose

membrane along with densitometric analysis of protein present was done to verify

equivalent protein loading between the lanes (data not shown). (*) indicates significant

difference from adult (6-month) value (P<0.05). (†) indicates significant difference from

age matched 33-month control animals (P<0.05). n = 6 for all groups.

86

Figure 3. 6. Acetaminophen treatment increases mTOR phosphorylation in aged cardiac

muscle. Protein isolates of hearts from 6-month contol, 33-month control and 33-month

treated hearts were analyzed by immunoblotting to determine the amount of total and

phosphorylated mTOR (Ser 2448). Ponceau S staining of the nitrocellulose membrane

along with densitometric analysis of protein present was done to verify equivalent protein

loading between the lanes (data not shown). (*) indicates significant difference from

adult (6-month) value (P<0.05). (†) indicates significant difference from age matched 33-

month control animals (P<0.05). n = 6 for all groups.

87

Figure 3. 7. Acetaminophen treatment increases ERK1/2 phosphorylation in aged cardiac

muscle. Protein isolates of hearts from 6-month contol, 33-month control and 33-month

treated hearts were analyzed by immunoblotting to determine the amount of total and

phosphorylated ERK1/2. Ponceau S staining of the nitrocellulose membrane along with

densitometric analysis of protein present was done to verify equivalent protein loading

between the lanes (data not shown). (*) indicates significant difference from adult (6-

month) value (P<0.05). (†) indicates significant difference from age matched 33-month

control animals (P<0.05). n = 6 for all groups.

88

Figure 3. 8. Acetaminophen treatment increases p70S6K phosphorylation in aged cardiac

muscle. Protein isolates of hearts from 6-month contol, 33-month control and 33-month

treated hearts were analyzed by immunoblotting to determine the amount of total and

phosphorylated p70S6K (Thr421/Ser424). Ponceau S staining of the nitrocellulose

membrane along with densitometric analysis of protein present was done to verify

equivalent protein loading between the lanes (data not shown). (*) indicates significant

difference from adult (6-month) value (P<0.05). (†) indicates significant difference from

age matched 33-month control animals (P<0.05). n = 6 for all groups.

89

Figure 3. 9. Acetaminophen treatment increases GSK-3β phosphorylation in aged cardiac

muscle. Protein isolates of hearts from 6-month contol, 33-month control and 33-month

treated hearts were analyzed by immunoblotting to determine the amount of total and

phosphorylated GSK-3β. Ponceau S staining of the nitrocellulose membrane along with

densitometric analysis of protein present was done to verify equivalent protein loading

between the lanes (data not shown). (†) indicates significant difference from age matched

33-month control animals (P<0.05). n = 6 for all groups.

90

Figure 3. 10. Proposed scheme for the effects of chronic acetaminophen treatment in

attenuating aging-associated cardic ROS and apoptosis. Recent studies indicated that the

aging associated increases in oxidative stress play a key role in cardiomyocyte apoptosis.

Chronic acetaminophen treatment can attenuate excess ROS leading to diminished

cardiomyocyte apoptosis through changes in Bax/Bcl2 ratio, caspase activation and by

regulating Akt phosphorylation. Note: Solid red arrow indicates increase and solid blue

arrow indicates decrease.

91

PAPER 3

The following paper corresponds to the specific aim III

92

Anti-arrhythmic effect of chronic acetaminophen treatment in the aging F344XBN

rat involves diminished myocardial fibrosis and altered miRNAs regulation

Sunil K. Kakarla1,2

, Srininvas Thulluri1, Nandini D.P.K. Manne

1,3, Kevin M. Rice

1,3,4,5,6,

and Eric R. Blough1,2,3,4, 5,6

1 Center for Diagnostic Nanosystems, Marshall University, Huntington, WV

2 Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of

Medicine, Marshall University, Huntington, WV 3 Department of Biological Sciences, Marshall University, Huntington, WV

4 Department of Internal Medicine, Joan C. Edwards School of Medicine, Marshall

University, Huntington, WV 5

Department of Exercise Science, Sport and Recreation, College of Education and

Human Services, Marshall University, Huntington, WV 6 Department of Biology, West Virginia State University, Charleston, WV

Running title: Acetaminophen confers cardioprotection in old rats

Keywords: acetaminophen; aging; arrhythmias; fibrosis; Cx43; miRNA

93

Abstract

Background: There is a growing need for pharmacological agents to manage

cardiovascular disease in the rapidly increasing elderly population. Acetaminophen has

been shown to exhibit cardioprotective effects following ischemia-reperfusion injury by

acting as an antioxidant. Recent data suggests that chronic acetaminophen ingestion

significantly attenuates age-associated increases in cardiac ROS and apoptosis. Methods

and results: Here, we examine the effect of chronic acetaminophen treatment on the

incidence of cardiac arrhythmias, Cx43 expression, myocardial fibrosis and miRNA

expression in the aging Fischer344XBrown Norway (F344XBN) rat hearts. Aging male

F344XBN rats (27-mo; n=6) were treated with acetaminophen (30mg/kg/day p.o.) for six

months and underwent serial electrocardiography before and after the completion of the

study. Aging was associated with a higher incidence of premature atrial and ventricular

arrhythmias, greater myocardial fibrosis and diminished localization of the gap junction

protein Cx43, all of which were diminished with chronic acetaminophen ingestion. The

expression of miR-1, miR-133, miR-214, miR-30a and miR-30d was 66-, 64-, 54-, 73-

and 74-% lower, respectively, in 33-month control animals compared to that of 6-month

animals (P < 0.05). Acetaminophen treatment significantly increased the expression of

miR-1 and miR-214, while the other miRNA remain unchanged. Conclusion: These

results indicate that acetaminophen may prevent the incidence of age-associated

arrhythmias and that this alteration is associated with diminished fibrosis and changes in

cardiac miRNA expression.

94

Introduction

Cardiovascular disease remains the foremost cause of death and morbidity

worldwide and is especially prevalent in the elderly. Nearly half the deaths occurring

from cardiovascular causes take place suddenly due to fatal ventricular arrhythmias

[267]. Aging by itself represents a major risk factor for cardiovascular disease and is

associated with structural and functional alterations in the cardiovascular system

including increased LV wall thickness, increased LV mass, impaired LV ejection and a

higher incidence of arrhythmias [30, 32]. Echocardiographic studies have demonstrated

that aging in the Fischer 344/NNiaHSd X Brown Norway/BiNia (F344XBN) rat is

characterized by age-associated alterations in cardiac structure and function [16, 17],

while other work has demonstrated that cardiac aging in these animals is associated with

increases in the amount of cardiac reactive oxygen species (ROS) [18, 268] and myocyte

apoptosis [20]. It has also been established that myocardial aging is characterized by LV

LV fibrosis and a reduction in cardiomyocyte number, which together act to decrease

myocardial compliance and increase the risk of developing ventricular dysfunction and

arrhythmias [20, 85].

The factors that regulate age-associated cardiac remodeling have not been

elucidated. The “free radical theory of aging” proposes that aging is characterized by the

gradual accumulation of damage to biomolecules by reactive oxygen species (ROS) [31].

Why ROS levels may be altered with aging is not fully understood, but it is thought that

these changes may be due, at least in part, to altered mitochondrial electron transport

leakage [269], changes in the activity of xanthine oxidase [270],

variation in the

expression of nitric oxide synthase [270], or increases in the amount of NADPH oxidases

95

[271]. It has been hypothesized that elevations in tissue ROS levels with age can

influence several cellular processes including cardiac hypertrophy, the structure and

function of blood vessels, inflammation, apoptosis and alteration of the extracellular

matrix profile [61, 64]. Whether interventions aimed at decreasing age-related increases

in ROS may be beneficial for the treatment of age-associated cardiac arrhythmias or

dysfunction is not well understood [272].

The molecular mechanisms governing the cardiac remodeling are not well

understood. Recent data has suggested that miRNAs (miRNAs), small ~ 22-nucleotide

noncoding RNAs that inhibit transcription or translation by interacting with the 3′

untranslated region (3′UTR) of target mRNAs [156], may play a role in the regulation of

cardiac differentiation and disease [163]. In this context, several miRNAs, including

miR-1, 21, 29, 30, 195, 133, 206 and 208 appear to play an important role in the process

of cardiac remodeling, putatively by regulating changes in gene expression that

accompany pathological cardiac hypertrophy and contractile dysfunction [162, 164, 165,

170]. The factor(s) that regulate the expression of miRNA are not clear; however, recent

studies have suggested that alterations in cellular ROS levels may be involved [178].

Similarly, how aging may affect the expression of cardiac miRNA has not been

elucidated.

Acetaminophen is a potent antioxidant that has been used therapeutically for

several decades [22], and recently has been shown to exhibit cardioprotective properties

[21]. It is thought that acetaminophen can help protect cells and tissues from the

damaging effects of peroxynitrite, myeloperoxidase, cyclooxygenase, and other peroxides

96

[21]. Recent work has also suggested that acetaminophen exhibits potent cardioprotective

activity in preventing ROS-associated cardiac damage in the rat and dog heart [197].

Whether the chronic administration of acetaminophen is effective for the treatment of

age-associated cardiac remodeling is not well understood. Using the tissues from the

same animals employed in this study we have previously reported that chronic

acetaminophen ingestion significantly attenuates age-associated increases in cardiac ROS

and apoptosis [203]. Here we extend upon these studies and examined the effect of aging

and chronic low dose acetaminophen treatment on the expression of cardiac miRNA and

the development of cardiac arrhythmias. Our data suggest that aging in F344XBN rats is

associated with increased incidence of cardiac arrhythmias, lowered expression of gap

junction protein connexin-43 (Cx43), increased myocardial fibrosis and altered regulation

of miRNA. In addition, we also demonstrate that chronic acetaminophen ingestion is

efficacious in preventing the incidence of age-associated arrhythmias and that this

alteration is associated with diminished fibrosis and changes in cardiac miRNA

expression.

97

Materials and Methods

Animals

All procedures were performed in accordance with the Marshall University

Institutional Animal Care and Use Committee (IACUC) guidelines, using the criteria

outlined by the American Association of Laboratory Animal Care (AALAC). Fischer

344/NNiaHSD x Brown Norway/BiNia (F344XBN) rats aged 6 and 27 months, were

purchased from the National Institutes on Aging. Adult F344XBN rats at 27-months of

age were chosen for the study since they do not exhibit significant cardiovascular deficit

at this age and roughly coincide with humans in the 5th

decade of life [15]. Rats were

housed two per cage in an AAALAC approved vivarium with a 12 hour light-dark cycle

and temperature maintained at 22 ± 2°C. Animals were fed ad libitum and allowed to

acclimate for 2 weeks before the initiation of any treatment or procedures. All animals

were examined for precipitous weight loss, failure to thrive or unexpected gait alterations

and animals with apparent abnormalities or tumors were removed from the study.

Materials

Acetaminophen tablets or pure compound used in the study was provided by

McNeil Pharmaceuticals (Fort Washington, PA). Antibodies against Cx43 and rabbit IgG

antibodies were purchased from Cell Signaling Technology (Beverly, MA, USA). Precast

10% SDS-PAGE gels were procured from Cambrex Biosciences (Baltimore, MD, USA),

and enhanced chemiluminescence (ECL) Western blot detection reagent was acquired

from Amersham Biosciences (Piscataway, NJ, USA). Restore Western blot stripping

98

buffer was obtained from Pierce (Rockford, IL, USA) and 3T3 cell lysates were from

Santa Cruz Biotechnology. All other chemicals were purchased from Sigma (St Louis,

MO, USA).

Acetaminophen treatment, electrocardiography and tissue collection

F344XBN rats (27-month; n = 6) were given acetaminophen (30 mg / kg body

weight / day) for 6 months in drinking water. Rats were treated until the age of 33-

months, which roughly coincides with humans in their seventh decade of life. Age-

matched F344×BN rats were maintained as controls. Heart rate and cardiac rhythm were

measured before the commencement of treatment and at the end of the treatment using

needle electrodes under ketamine-xylazine (4:1) cocktail (50 mg/kg, I/P) anesthesia.

Tissues were collected under anesthesia using a ketamine-xylazine cocktail (40:10

mg/kg i.p.) and supplemented as necessary for reflexive response. After midline

laparotomy, the heart was removed and placed in Krebs-Ringer bicarbonate buffer (KRB)

containing; 118 mM NaCl, 4.7 mM KCl, 2.5 mM CaCl2, 1.2 mM KH2PO4, 1.2 mM

MgSO4, 24.2 mM NaHCO3, and 10mM α-D-glucose, (pH 7.4) equilibrated with 5% CO2

/ 95% O2 and maintained at 37°C. Isolated hearts were quickly massaged to remove any

blood from the ventricles, cleaned of connective tissue, weighed, and immediately snap

frozen in liquid nitrogen. Liver and kidneys were excised immediately after removing

the heart and either snap-frozen under liquid nitrogen or stored in 5% buffered

formaldehyde.

99

Histology

Tissue specimens were serially sectioned (6 μm) using an IEC Minotome cryostat

and collected on poly-lysine coated slides. After fixing in acetone, (-20 °C for 2 min)

sections were stained with hematoxylin and eosin and mounted. Morphometric evaluation

was performed with the use of a computerized imaging analysis system (Olympus

MicroSuite™ Basic). Masson’s trichrome staining of serially sectioned cardiac tissue was

performed according to the manufacturer’s guidelines (Poly Scientific R & D Corp., Bay

Shore, NY).

Immunohistochemistry

Immunostaining for Cx43 was visualized by immunofluorescence as outlined by

the antibody manufacturer. Briefly, sections were washed three times with phosphate-

buffered saline (PBS) containing 0.5% Tween-20 (PBS-T), pH 7.5. After incubation for

30 min in a blocking solution (3% BSA), sections were incubated with specific antisera

diluted in PSB-T (antibody dilution of 1:100) for 1 h at 24°C in a humidified chamber.

After washing three times with PBS, sections were incubated with FITC anti-rabbit IgG

(1:200) for 30 min at 24°C in a humidified chamber. DAPI was also included in the

secondary antibody solution at a concentration of 1.5 μg/ml in order to visualize cell

nuclei. After a final PBS wash and mounting, specimens were visualized by

epifluorescence using an Olympus fluorescence microscope (Melville, NY, USA) fitted

with a 40x objective. Images were recorded digitally using a CCD camera and analyzed

using Olympus MicroSuite™ Basic from Olympus America (Melville, NY, USA).

100

Immunoblot analysis

Tissues were pulverized in liquid nitrogen using a mortar and pestle until a fine

powder was obtained and washed three times with ice-cold PBS as described previously

[273]. Samples were then suspended on ice in 100 μl of TPER (2 ml/g tissue weight;

Pierce, Rockford, IL, USA) containing protease (P8340, Sigma-Aldrich, Inc., St. Louis,

MO, USA) and phosphatase inhibitors (P5726, Sigma-Aldrich, Inc., St. Louis, MO,

USA). After lysis, homogenates were centrifuged 10 min at 13,000 x g and the

supernatant collected. Protein concentrations of homogenates were determined in

triplicate via the Bradford method (Pierce) using bovine serum albumin as a standard.

Samples were diluted to a concentration of 2.5 mg/ml in SDS-loading buffer and boiled

for 5 min. Aliquots (50 μg of total protein for each sample) were separated on a 10%

SDS-PAGE gel. Transfer of protein onto nitrocellulose membranes was performed using

standard conditions. Membranes were stained with Ponceau S and the amount of protein

quantified by densitometric analysis to confirm successful transfer of proteins and equal

loading of lanes [20, 203]. Semiquantitiative immunodetection of specific proteins was

performed using densitometry. Immunoblots were stripped with Restore Western blot

stripping buffer as described by the manufacturer to obtain direct comparisons between

expression and phosphorylation levels of different signaling molecules. After verifying

the absence of residual HRP activity by treating the membrane with the ECL reagent,

membranes were washed and reprobed. Randomization of antibody incubation was

utilized to minimize potential experimental error associated with membrane stripping.

101

miRNA analysis

Total RNA was harvested from cardiac tissues using TRIzol Reagent,

(Invitrogen). Total RNA was solubilized in RNase-free H2O and quantified in duplicate

by measuring the optical density (OD) at 260 nm. Purity of RNA was assured by

obtaining an OD260/OD280 ratio of ∼2.0. Two micrograms of RNA were reverse

transcribed (RT) using the QuantiMiR RT kit (System Bioscience) according to standard

methods (Systems Bioscience). Control RT reactions were done in which the RT enzyme

was omitted. The control RT reactions were PCR amplified to ensure that DNA did not

contaminate the RNA. Cyber green-based real-time qPCR was performed by using a

7500 Real-Time PCR system (Applied Biosystems, Foster City, CA) and gene-specific

primers for the miRNA of interest designed according to the guidelines outlined in the

QuantiMiR RT kit (System Bioscience). Melt analysis was used after each PCR run to

ensure amplification of only a single product. All samples were normalized to an internal

control housekeeping gene (U1 small nuclear RNA (snRNA)) [274]. Relative fold

changes in miRNA within each group and basal differences between groups were

determined from the Ct values after normalization to their respective housekeeping genes

using the 2-∆ct

method [275].

Data Analysis

Results are presented as mean ± SEM. Data were analyzed by using the

SigmaStat 3.5 statistical program. A one-way analysis of variance was performed for

overall comparisons while the Student-Newman-Keuls post hoc test was used to

102

determine differences between groups. The level of significance accepted a priori was ≤

0.05.

Results

As reported previously [203], compared to 6-month animals, body weight was

significantly higher with age (6 C: 406 ± 15 vs. 33 C : 504 ± 23* vs. 33 T : 506 ± 13*;

(n=6; P < 0.05)). Chronic acetaminophen ingestion attenuated age-associated increases in

heart weight (6 C: 1.05 ± 0.04 vs. 33 C: 1.85 ± 0.06* vs. 33 T: 1.65 ± 0.05*†; (n=6; P <

0.05)) and heart / body weight ratio (6 C: 0.259 ± 0.01 vs. 33 C: 0.370 ± 0.02* vs. 33 T:

0.327 ± 0.01†; (n=6; P < 0.05)).

Aging is associated with increased incidence of cardiac arrhythmias

Confirming our previous observations [17], serial electrocardiography showed

evidence of increased incidence of arrhythmias with age (Figure 4.1). Chronic

acetaminophen treatment led to a marked decrease in the incidence of premature atrial

(PAC) and ventricular (PVC) contractions (Figures 4.1A, 4.1B).

Aging is associated with greater myocardial fibrosis

It is thought that the intrinsic properties of the myocardium change during aging

due to connective tissue alterations [276]. In agreement with this possibility, we found

increased perivascular fibrosis as well as subcellular alterations of the cardiomyocytes

and their junctions in the left ventricles of the aged control animals suggesting a

103

deterioration of heart function. Similarly, qualitative histological analysis on heart

sections of 33-month acetaminophen treated rats revealed the presence of fibrosis with

the incidence of these findings appearing to be less than that observed in the untreated

control animals (Figure 4.2).

Aging is associated with diminished connexin-43 levels

Compared to 6-month animals, Cx43 expression in the 33-month control animals

was 25% lower (P < 0.05) (Figure 4.3A). Acetaminophen treatment for 6-months

significantly increased Cx43 levels by 8% (P < 0.05) (Figure 4.3A). These results were

corroborated by immunohistochemical analysis of these hearts. Consistent with previous

findings [277], gap junctional Cx43 immunoreactivity was primarily observed at the

intercalated discs (Figure 4.3B). When compared to hearts obtained from 6-month rats,

Cx43 immunoreactivity at the intercalated disc regions of the membrane was markedly

lower in 33-mo control rat hearts, and cytosolic Cx43 immunoreactivity appeared to be

clustered inside the fibers of old hearts (Figure 4.3B).

Aging is associated with alterations in cardiac miRNA expression

In order to evaluate whether miRNAs play a significant role in the aging process,

a highly sensitive, semi-quantitative RT-PCR method was employed to analyze the

expression of miRNA thought to be involved in cardiac remodeling. Compared to 6-

month animals, the amount of miR-1, miR-133, miR-214, miR-30a and miR-30d was 66-

, 64-, 54-, 73- and 74-% lower, respectively in 33-month control animals (Figures 4.4-

104

4.6). The amount of miR-206, miR-21, miR-24, miR-195 and miR-29a was not altered

with aging (data not shown).

Acetaminophen treatment is associated with alterations in miRNA expression

Compared to 33-month control animals, acetaminophen treatment increased the

expression of miR-1 and miR-214 expression levels by 29-, and 53 %, respectively, (P <

0.05) (Figures 4.4 and 4.6). The amount of miR-133, miR-30a and miR-30d did not

change with treatment.

105

Discussion

Multiple experimental models have been used to assess the effects of aging on

ROS and CVD, including dogs, the Fisher 344 (F344) and Fischer 344/NNiaHSd X

Brown Norway/BiNia (F344XBN) aging rat models, and other species [278]. A recent

review of aging rat models concluded that the F344XBN rat strain seems to exhibit

“physiological aging” to a greater degree than others [13]. Indeed, previous studies have

demonstrated that aging in the F344XBN rats is characterized by higher cardiac ROS [18,

203] and apoptosis [20]. Other data have suggested that acetaminophen may function as a

potent antioxidant [27, 29]. Given the possible linkage between higher cardiac ROS and

cardiac dysfunction, we postulated that the chronic ingestion of acetaminophen would

attenuate detrimental changes in the aging F344XBN heart. The main findings of this

study were that chronic acetaminophen ingestion is capable of decreasing age-associated

increases in cardiac arrhythmias and that aging in the F344XBN heart is associated with

changes in miRNA expression.

Aging in the F344XBN heart is associated with greater incidence of cardiac

arrhythmias

Nearly half the deaths occurring from cardiovascular causes take place suddenly

due to fatal ventricular arrhythmias [267]. Similar to previous studies, we observed an

age-associated increase in the incidence of ventricular arrhythmias in the F344XBN rat

[16-18]. Of interest is our finding that 6-months of acetaminophen treatment appeared to

decrease the incidence of PAC and PVC (Figure 4.1). Recent data has implicated a link

106

between the loss of the gap junction protein connexin-43 (Cx43) and fatal arrhythmias.

For example, using a heart-specific Cx43 conditional knockout mouse model in which

cardiac Cx43 abundance decreases rapidly within the first few weeks of age, Danik and

colleagues demonstrated a decrease in cardiac conduction velocity and a dramatic

increase in susceptibility to inducible lethal ventricular tachyarrhythmias [279]. To

ascertain if alterations in Cx43 levels exist in the aging F344XBN heart we examined the

regulation of Cx43 using immunoblot analysis. Consistent with our electrocardiography

data, we observed that the expression of Cx43 is significantly lowered with aging and

importantly, that acetaminophen treatment is associated with increased protein levels of

Cx43 (Figure 4.3A). Immunohistochemical analysis revealed that the distribution of

Cx43 appears to be morphologically well preserved at the intercalated discs in the young

6-month animals. With increasing age, the amount of Cx43 immunoreactivity appeared to

decrease markedly at the intercalated disc regions of the 33-month control rat hearts

(Figure 4.3B).

Although these results, taken together, suggest that chronic acetaminophen

ingestion may be able to influence the expression of Cx43 it is unlikely that the

magnitude of the differences we see in the regulation of Cx43 can, by itself, explain the

decrease in age-associated arrhythmias we observed in the acetaminophen treated

animals. Indeed, previous data has suggested that Cx43 protein losses of up to ~40% are

insufficient by themselves to induce cardiac arrhythmias [279]. How acetaminophen may

decrease cardiac arrhythmias in the aging F344XBN heart is presently unclear. Given the

putative link between cardiac ROS levels and arrhythmias [248], it is likely that the anti-

107

arrhythmic effects of acetaminophen are related to its ability to act as an anti-oxidant [27,

29]. Using the same heart tissues from the present study, we have previously reported that

chronic acetaminophen ingestion significantly diminished age-associated increases in

cardiac ROS and apoptosis in the F344XBN rats [203]. Whether the changes we see in

Cx43 expression with acetaminophen treatment are directly linked to acetaminophen-

associated changes in cardiac ROS levels is unclear and beyond the scope of this study.

Nonetheless, these data are consistent with the notion that acetaminophen is capable of

increasing Cx43 expression and that long term acetaminophen ingestion may be

beneficial in reducing the incidence of age-related arrhythmias.

Using the F344XBN aging rat model, we have previously reported that aging is

associated with higher nitrosative and oxidative stress [18] ultimately leading to greater

cardiomyocyte apoptosis [20]. Other studies have also shown that myocardial aging is

characterized by LV fibrosis that has been linked to a progressive reduction in

cardiomyocyte number, increases in cardiac fibroblast (CF) proliferation, and the

deposition of collagen in the LV [85, 280]. Excessive fibrosis can cause decreases in

myocardial compliance and may be linked to the development of ventricular dysfunction

and arrhythmias [139]. Likewise, our findings also show significantly higher fibrosis in

33-month control animals compared to 6-month animals (Figure 4.2). Interestingly, six

months of chronic acetaminophen ingestion significantly diminished myocardial fibrosis

compared to that observed in age-matched control animals (Figure 4.2). These results are

in agreement with our previous report suggesting that chronic acetaminophen treatment

significantly diminishes age-related cardiomyocyte apoptosis in male F344XBN rats

[203]. Whether the decrease in cardiac fibrosis we see in acetaminophen treated aged rats

108

is linked to decreases in the amount of cardiomyocyte apoptosis or due to alterations in

signaling pathways that control cardiac fibrosis warrants further investigation. Similarly,

whether the decrease in cardiac fibrosis we observed with acetaminophen treatment is

directly responsible for the diminished cardiac arrhythmias we observe in the treated rats

is currently not clear.

Aging in the F344XBN heart is associated with diminished miR-1, miR-133, miR-

214, miR-30a and miR-30d expression

Previous work by our laboratory and others have demonstrated that aging in the

male F344XBN is associated with increases in heart mass, LV hypertrophy, increased

incidence of cardiac arrhythmias, decreased ventricular compliance and impairment in

systolic and diastolic function [16, 17] as well as increased expression of oxidative-

nitrosative stress [18] and cardiomyocyte apoptosis [20]. Using semi-quantitative real

time RT-PCR we demonstrate a lower amount of miR-1, miR-133, miR-214, miR-30a

and miR-30d with aging (Figures 4.4-4.6). The physiological significance of these

findings awaits further experimentation. Previous data has suggested that miR-1 and

miR-133 expression is upregulated, downregulated, and unchanged during cardiac

remodeling [164-166]. Why aging may decrease the expression of miR-1, miR-133,

miR-214, miR-30a and miR-30d levels is not fully understood. The reasons for divergent

results is unknown but may be related to differences in disease models, extent of disease,

or the myocardial regions sampled.

Recent reports have implicated miR-1 as participating in the etiology of

ventricular arrhythmias as it is thought that this miRNA post-transcriptionally represses

109

the expression of Cx43 [166]. Interestingly, the increase in arrhythmias we observe with

aging were associated with decreases in Cx43 (Figure 4.3), and decreases in miR-1

expression (Figure 4.4). This result is in opposition to the previous findings suggesting

that decreases in miR-1 expression will lead to increase in Cx43 expression. Indeed, the

role that miR-1 plays in regulating Cx43 in the aging heart is still unclear as treatment

associated increases in miR-1 expression (Figure 4.4) would lead one to predict that Cx43

levels would be decreased. Clearly this postulate was opposite to what we found as

acetaminophen treatment led to slight yet significant increases in Cx43 expression. In

addition to its role in regulating Cx43, it is also possible that miR-1 acts as an inhibitor of

cardiac hypertrophy. For example, the expression of miR-1 in culture inhibits serum-

induced hypertrophy and is associated with inhibited RasGAP, Cdk9 and fibronectin

expression [281]. Recent data has also shown that cardiac-specific over expression of

miR-1 in the embryonic heart inhibits cardiomyocyte proliferation and prevents

expansion of the ventricular myocardium [168]. As such, it is possible that the decrease

in miR-1 we see with aging and increase in the aged acetaminophen treated animals may

indicate a role for miR-1 to limit cardiac remodeling. The mechanism(s) underlying these

alterations is currently unclear but may be related to the antioxidant properties of

acetaminophen. Indeed, recent experiments have shown that the expression of several

miRNA species can be modulated by changes in cellular ROS [282]. For example, the

expression of miR-21 is increased following the exposure of cultured myocytes to

increased hydrogen peroxide while miR23b is down regulated in vascular smooth muscle

cells after exposure to hydrogen peroxide [283]. Further experiments perhaps using other

methods, pharmacological treatments or time points of observation will no doubt be

110

useful in increasing our understanding of how ROS levels may influence miRNA

expression.

Similar to many micro-RNAs, the role of miR-133 in cardiac remodeling has yet

to be established. Recent work has demonstrated that increased miR-133 results in the

suppression of protein synthesis and inhibition of hypertrophic growth in cultured

neonatal cardiac myocytes [164]. In this same study the authors also showed that

antisense RNA oligonucleotides targeting miR-133 resulted in cardiac hypertrophy and

reinduction of the fetal gene program [164]. Other studies have suggested that miR-133

may act as antiapoptotic factor by repressing the translation of caspase-9 [172]. Given

these postulates it is possible that the age-associated decrease in miR-133 we observe

may play a role in the ventricular hypertrophy seen in these animals with advanced age.

Alternatively, miR-133 has also been implicated in the pathogenesis of cardiac

arrhythmias as it has been shown that this molecule can regulate the ether-a-go-go related

gene (ERG), which encodes a key portion of the cardiac potassium channel [284].

Whether the changes in miR-133 we observe are related to factors controlling cardiac

hypertrophy or cardiac rhythm will require further investigation.

In addition to miR-1 and miR-133, recent studies have also implicated a role for

miR-30 in cardiac remodeling. Liu et al. showed that miR-133 knockout mice develop

severe fibrosis and heart failure [285]. Recently, Duisters et al. investigated the

regulation of connective tissue growth factor (CTGF), and showed that the expression of

miR-133 and miR-30 was inversely related to the amount of CTGF in rodent models of

heart disease and in human patients with pathological LV hypertrophy [176]. Similar to

their findings, the age-related increase in fibrosis we observe in F344XBN rat hearts are

111

associated with diminished expression of miR-133, miR-30a and miR-30d (Figures 4.4,

4.5). Conversely, the diminished myocardial fibrosis we see in acetaminophen treated

aged rats was not associated with any alterations in the expression of miR-133, miR-30a

and miR-30d.

The role of miR-214 in cardiac remodeling has not been elucidated. Recent study

suggests that miR-214 is upregulated during pathological hypertrophy; however,

transgenic mice over-expressing miR-214 exhibited no cardiac abnormalities [162]. Our

results indicate that miR-214 expression significantly decreases with age in aged control

rats and that the expression of this molecule remains unchanged with acetaminophen

ingestion (Figure 4.6). Understanding why the expression of miR-214 is altered

differently in aged hypertrophied hearts compared to other models of pathological

hypertrophy remains unclear and will require further investigation.

In conclusion, the results of this study show that chronic acetaminophen treatment

decreases the incidence of age-associated arrhythmias and that this alteration is

associated with increased Cx43, diminished myocardial fibrosis and alterations in cardiac

miRNA expression. Given that acetaminophen is well tolerated in humans and approved

by the FDA for over the counter sale, the possibility exists that this widely available drug

may be an attractive therapeutic option in the war against heart disease. Further

experiments using different animals and methods may be warranted to expand upon these

observations.

112

Funding

This study was supported by funding from McNeil Pharmaceuticals and the Department

of Energy Grant DE-PS02-09ER09-01 to ERB.

113

Figures

Figure 4. 1. Chronic acetaminophen treatment decreases incidence of cardiac

arrhythmias. (A) Percentage incidence of premature atrial (PAC) and ventricular (PVC)

contractions observed in the 33-mo control and 33-mo APAP-treated rats (n=6). (B)

Representative echocardiographs (Lead II) recorded from 6-mo control, 33-mo control

and 33-mo APAP-treated rats (n=6).

114

Figure 4. 2. Chronic acetaminophen attenuates myocardial fibrosis in aged rat hearts.

Representative ventricular myocardium sections from 6-mo control, 33-mo control and

33-mo treated rats treated with Masson’s Trichrome stain (A) and Hematoxylin & Eosin

stain (B) (n=4).

115

Figure 4. 3. Chronic acetaminophen preserves myocardial membrane Cx43. (A) Protein

isolates of hearts from 6-month control, 33-month control and 33-month treated hearts

were analyzed by immunoblotting to determine Cx43 protein levels. Ponceau S staining

of the nitrocellulose membrane along with densitometric analysis of protein present was

done to verify equivalent protein loading between the lanes. *Significant difference from

adult (6-month) value (P < 0.05). †Significant difference from age matched 33-month

control animals (P < 0.05). n = 6 for all groups (B) Representative immunohistochemical

images of hearts sections obtained from 6-mo control, 33-mo control and 33-mo APAP-

treated rat hearts using an antibody against Cx43.n=4 hearts per age group

116

Figure 4. 4. Aging decreases myocardial miR-1 and miR-133 expression. Quantitative

RT-PCR analyses of miRNA expression in 6-mo control, 33-mo control and 33-mo

treated rat hearts using SYBR green I. Relative expression of miR-1 (A) and miR-133 (B)

between groups was calculated after normalization to U1 miR expression in the same

samples. * Significantly different from 6-mo Control. † Significantly different from 33-

mo control. For all comparisons, P < 0.05; n = 4

117

Figure 4. 5. Aging decreases myocardial miR-30a and miR-30d expression. Quantitative

RT-PCR analyses of miRNA expression in 6-mo control, 33-mo control and 33-mo

treated rat hearts using SYBR green I. Relative expression of miR-30a (A) and miR-30d

(B) between groups was calculated after normalization to U1 miR expression in the same

samples. * Significantly different from 6-mo Control. † Significantly different from 33-

mo control. For all comparisons, P < 0.05; n = 4

118

Figure 4. 6. Chronic acetaminophen treatment alters myocardial miR-214 expression.

Quantitative RT-PCR analyses of miRNA expression in 6-mo control, 33-mo control and

33-mo treated rat hearts using SYBR green I. Relative expression of miR-214 between

groups was calculated after normalization to U1 miR expression in the same samples. *

Significantly different from 6-mo Control. † Significantly different from 33-mo control.

For all comparisons, P < 0.05; n = 4

119

CHAPTER IV

GENERAL DISCUSSION

The primary purpose of this study was to investigate the molecular mechanisms

underlying age-associated alterations in the structure and function of the aging F344XBN

rat heart. In addition, we also examined the efficacy of chronic acetaminophen ingestion

to prevent or reverse age-associated cardiac dysfunction. To accomplish these goals, we

determined the effects of aging and acetaminophen ingestion on heart size, mass,

myocyte cross sectional area, myocyte histology, indices of oxidative stress, the

incidence of cardiac apoptosis, protein expression and cardiac structure and function.

The aged (65+ years of age) now comprise the most rapidly growing segment of

the United States population and mounting evidence suggests that advancing age is

associated with increased risk of cardiovascular disease and a progressive decline in

cardiac function [3, 30]. It is well established that aging is associated with myocardial

structural changes including loss of myocytes, hypertrophy of the remaining

cardiomyocytes with progressive increase in myocyte cell volume per nucleus, reduction

in the number of conduction cells and wall thickening [1]. The exact molecular

mechanisms underlying the structural and functional alterations that lead to increased

susceptibility of the aging heart to fatal arrhythmias are not well understood. Age-related

alterations in cardiac structure and function have been mostly studied in human hearts at

the time of transplantation, while animal models have been found to be extremely useful

for the assessment of pathological processes and the development of new treatments [13].

A recent review of aging rat models concluded that the F344XBN rat strain seems to live

120

longer while also exhibiting “physiological aging” to a greater degree than other rat

models [13-15].

Age-related alterations in cardiac structure and function of F344XBN rats

Recent investigations using the Fischer F344XBN rat model have demonstrated

that aging in these animals is associated with an increased incidence of cardiac

arrhythmias as well as impairment of systolic and diastolic function [16, 17]. Hacker et

al. performed echocardiographic and hemodynamic analyses in male F344XBN rats from

adulthood to the very aged (n = 6 per 12-, 18-, 21-, 24-, 27-, 30-, 33-, 36-, and 39-mo-old

group) and revealed that aged rats had an increased LV mass-to-body weight ratio,

deterioration of systolic function, gradual increase in fibrosis and a decrease in

cardiomyocyte volume density [16]. In an another study, Walker et al. performed two-

dimensional ECHO measurements on male F344/BN rats aged 6-, 30-, and 36-month

and demonstrated that aging was associated with cardiac arrhythmias in 72% of 36-

month rats, 10% of 30-month rats and 0% in 6-month rats [17]. Previous work by our

laboratory has shown that aging in the F344XBN heart is characterized by increased

oxidative-nitrosative stress and that these findings were highly correlated with increases

in LV wall thickness [18]. Furthermore, this study has also demonstrated that age-related

increases in cardiac oxidative-nitrosative stress were significantly correlated with

changes in the expression and/or regulation of proteins involved in transcriptional (NF-

κB) activities, signaling (MAPKs along with Src), apoptotic (Bcl-2, Traf-2), and cellular

stress (HSPs) [18]. Other work has shown that aging in the F344XBN heart is associated

121

with increases in cardiac apoptosis and that this increase in cell death may be due, at least

in part, to elevations in cardiac ROS [20].

Findings from the present investigation appear to confirm that aging in F344XBN

rat heart is associated with increases in cardiac ROS, apoptosis and fibrosis ultimately

leading to an increased incidence of arrhythmias. In addition, our findings from this study

also demonstrate that aging in F344XBN rats is associated with significant increase in

heart size, heart weight, heart weight to body weight ratio, and cardiomyocyte fiber cross

sectional area. Specifically, we observed that heart size and body weight gradually

increased with age but that the heart to body weight ratio remain unchanged up to 30

months before increasing significantly at 36-months of age. In a similar fashion, the

cardiomyocyte fiber cross sectional areas tended to increase rapidly from 30 – 36 months

before reaching a level at 36 months that was nearly three times larger than that observed

in the 6-month adult animals. Furthermore, we also report that very aged hearts exhibit

the activation of multiple signaling mechanisms during the hypertrophic response. Our

findings suggest that age-related cardiac hypertrophy in F344XBN rats is predominantly

mediated via activation of the ERK1/2 and Akt signaling pathways. We observed

increased phosphorylation (activation) of ERK1/2, Akt at Ser 473 and Thr 308 in both the

33- and 36-month rat hearts. This activation of Akt was confirmed when we examined

the phosphorylation of Akt substrate, GSK3β. Likewise, PTEN, a negative modulator of

PI3K/Akt/mTOR pathway [225] that is inactivated upon phosphorylation, was found to

be significantly higher in the very aged 33- and 36-month animals. Like Akt, a growing

body of evidence suggests that mTOR may play a key role in regulating cell growth and

122

protein synthesis [129, 226]. Our findings also demonstrate a large increase in the amount

of mTOR phosphorylation in the very aged 36-month animals. It is thought that mTOR

may be involved in controlling a number of downstream signaling proteins that regulate

mRNA translation [133] such as the translational repressor proteins 4E-binding protein-1

(4E-BP1) and the ribosomal protein S6 kinase (rp6k) [123]. Consistent with our findings

of an increased mTOR phosphorylation, we observed that the phosphorylation of 4EBP1

and rpS6 was increased significantly in the very aged 36-month animals. Taken together,

our current and previous findings [17, 18, 20, 203] suggest that aging in the F344XBN rat

heart is associated with diminished cardiac function, elevations in oxidative stress,

increased cardiomyocyte apoptosis and cardiomyocyte hypertrophy with the latter

occurring probably as a result of increased activation of the ERK1/2 and Akt signaling

pathways.

Cardioprotective properties of acetaminophen in aged F344XBN rats

Acetaminophen exhibits antipyretic and analgesic activities and has been widely

used over the past several decades. Being a phenolic compound, acetaminophen has an

inherent ability to scavenge free radicals. A recent study has suggested that this

compound may be an effective antioxidant given its ability to protect against membrane

lipid oxidation through the scavenging of peroxyl radicals and peroxynitrite [192].

Merrill et al. have shown that acetaminophen protects the isolated guinea pig hearts from

the damaging effects of myocardial ischemia and reperfusion, that it preserves

myofibrillar ultrastructure, and is associated with the attenuated production of both

hydroxyl radicals and peroxynitrite [29]. In an another study, Merrill et al. investigated

123

the effect of acetaminophen on pentobarbital pretreated isolated guinea pig hearts that

underwent ischemia-reperfusion and reported that acetaminophen (0.35mmol/l)

significantly reduced the frequency of pentobarbital induced arrhythmias [27].

Collectively these findings suggest that under selective conditions (e.g., those causing

release of free radicals and other oxidants) acetaminophen can act as an antioxidant and

that this agent is able to prevent ventricular arrhythmias [27, 29]. Chronically

administered acetaminophen has also been shown to provide cardioprotection to the

postischemic, reperfused rodent myocardium [28]. Here for the first time, we have

investigated the efficacy of chronic acetaminophen ingestion to prevent or reverse the

oxidative-nitrosative stress mediated alterations in cardiac structure and function in aged

F344XBN rats.

Our findings suggest that acetaminophen is able to diminish age-related increases

in cardiac reactive oxygen species (ROS) and apoptosis in these animals. Compared to 6-

month control animals, indices of oxidative (superoxide anion [O2•–] and 4-hydroxy-2-

nonenal [4-HNE]) and nitrosative (protein nitrotyrosylation) stress were markedly

increased in 33-month old rat hearts. Long term treatment with acetaminophen

(30mg/kg/day p.o. for six months) exhibited diminished age-related increases in cardiac

ROS levels and TUNEL positive nuclei, and these changes were accompanied by

improvements in the Bax/Bcl2 ratio, diminished evidence of caspase-3 activation and

increased phosphorylation of protein kinase B (Akt), ERK1/2, p70S6K and GSK-3β.

How acetaminophen might decrease cardiac ROS is not well understood at this time and

warrants further investigation. Previous reports suggest that acetaminophen may act as an

antioxidant as it is a mono-phenol [23]. Improving our understanding of these

124

mechanisms and how they may interact with other drugs and disease processes could lead

to new ways in improving our ability to manage heart disease.

In addition, we have also examined the efficacy of chronic acetaminophen

treatment on the incidence of cardiac arrhythmias, Cx43 expression, myocardial fibrosis

and miRNA expression in the aging F344XBN rats. In agreement with our previous

findings [17], serial electrocardiography showed evidence of increased incidence of

arrhythmias with age. Importantly, chronic acetaminophen treatment led to a marked

decrease in the incidence of premature atrial (PAC) and ventricular (PVC) contractions.

Recent data has implicated a link between the loss of the gap junction protein Cx43 and

fatal arrhythmias. For example, using a heart-specific Cx43 conditional knockout mouse

model in which cardiac Cx43 abundance decreases rapidly within the first few weeks of

age, Danik and colleagues have demonstrated a decrease in cardiac conduction velocity

and a dramatic increase in susceptibility to inducible lethal ventricular tachyarrhythmias

[279]. Here, we observed that the expression of Cx43 is significantly lowered with aging

and importantly that acetaminophen treatment is associated with increased protein levels

of Cx43. Although these findings, taken together, suggest that chronic acetaminophen

ingestion may be able to influence the expression of Cx43, it is unlikely that the

magnitude of the differences we see in the regulation of Cx43 can, by itself, explain the

decrease in age-associated arrhythmias we observed in the acetaminophen treated

animals. Indeed, previous data have suggested that Cx43 protein losses of up to ~40% are

insufficient by themselves to induce cardiac arrhythmias [279]. How acetaminophen may

decrease cardiac arrhythmias in the aging F344XBN heart is presently unclear. Given the

putative link between cardiac ROS levels and arrhythmias [248], it is likely that the anti-

125

arrhythmic effects of acetaminophen are related to its ability to act as an anti-oxidant [27,

29]. Whether the changes we see in Cx43 expression with acetaminophen treatment are

directly linked to acetaminophen-associated changes in cardiac ROS levels is unclear and

beyond the scope of this study.

Previous studies have demonstrated that myocardial aging is characterized by LV

LV fibrosis that has been linked to a progressive reduction in cardiomyocyte number,

increases in cardiac fibroblast (CF) proliferation, and the deposition of collagen in the LV

[85, 280]. Excessive fibrosis can cause decreases in myocardial compliance and may be

linked to the development of ventricular dysfunction and arrhythmias [139]. Here, we

also observe significantly higher fibrosis in aged F344XBN rat hearts and, interestingly,

that six months of chronic acetaminophen ingestion significantly diminished myocardial

fibrosis, compared to that observed in age-matched control animals. These results are in

agreement with our previous findings suggesting that chronic acetaminophen treatment

significantly diminishes age-related cardiomyocyte apoptosis in male F344XBN rats.

Whether the decrease in cardiac fibrosis we see in acetaminophen treated aged rats is

linked to decreases in the amount of cardiomyocyte apoptosis or due to alterations in

signaling pathways that control cardiac fibrosis warrants further investigation. Similarly,

whether the decrease in cardiac fibrosis we observed with acetaminophen treatment is

directly responsible for the diminished cardiac arrhythmias we observe in the treated rats

is currently not clear. In summary, our findings suggest that chronic acetaminophen

treatment in F344XBN rats decreases age-related increases in cardiac ROS, the incidence

of age-associated arrhythmias, increased Cx43 protein expression, and diminished

evidence of myocardial fibrosis.

126

Regulation of miRNA with aging and acetaminophen treatment in F344XBN rats

MiRNAs (miRNAs) are small ~ 22-nucleotide noncoding RNAs that inhibit

transcription or translation by interacting with the 3′ untranslated region (3′UTR) of

target mRNAs [156]. Recent data has suggested that miRNA may play a role in the

regulation of cardiac differentiation and disease [163]. In this context, several miRNAs

including miR-1, 21, 29, 30, 195, 133, 206 and 208 have been described as playing an

important role in the process of cardiac remodeling, possibly by regulating changes in

gene expression that accompany pathological cardiac hypertrophy and contractile

dysfunction [162, 164, 165, 170]. The factors that regulate the expression of miRNA are

not clear although recent studies have suggested that alterations in cellular ROS levels

may be involved [178]. Similarly, how aging may affect the expression of cardiac

miRNA and the effect of chronic use of antioxidants have not been elucidated. Here, for

the first time, we have investigated the regulation of miRNA and the alterations with

chronic acetaminophen ingestion in aged F344XBN rat hearts. Our findings suggest that

aging in F344XBN rat hearts is associated with a lower amount of miR-1, miR-133, miR-

214, miR-30a and miR-30d. Acetaminophen treatment increased the expression of miR-1

and miR-214, while the amount of miR-133, miR-30a and miR-30d remain unchanged.

The physiological significance of these findings awaits further experimentation. Previous

data have suggested that miR-1 and miR-133 expression is upregulated, downregulated,

and unchanged during cardiac remodeling [164-166]. Why aging may decrease the

expression of miR-1, miR-133, miR-214, miR-30a and miR-30d levels is not fully

understood. The reasons for divergent results is unknown but may be related to

differences in disease models, extent of disease, or the myocardial regions sampled.

127

SUMMARY

1. Aging in F344XBN rats is associated with significant increases in heart size, heart

weight, heart weight to body weight ratio and cardiomyocyte fiber cross sectional

area.

2. Aging associated cardiac hypertrophy in F344XBN rats is predominantly regulated

via activation of the ERK1/2 and Akt signaling pathways.

3. Age-related cardiac hypertrophy in F344XBN rats is associated with increased

protein translational signaling.

4. Chronic acetaminophen ingestion diminished age-associated increases in indices of

oxidative (superoxide anion [O2•–] and 4-hydroxy-2-nonenal [4-HNE]) and

nitrosative (protein nitrotyrosylation) stress in F344XBN rat hearts.

5. Acetaminophen treated animals exhibited diminished age-related increases in cardiac

TUNEL positive nuclei, improvements in the Bax/Bcl2 ratio, and diminished

evidence of caspase-3 activation.

6. Acetaminophen increased phosphorylation of Akt, ERK1/2 and GSK-3β which play a

key role in cell survival signaling.

7. Aging in F344XBN rats is associated with increased incidence of cardiac

arrhythmias, lowered expression of gap junction protein connexin-43 (Cx43),

increased myocardial fibrosis and altered regulation of miRNA.

8. Chronic acetaminophen ingestion is efficacious in preventing the incidence of age-

associated arrhythmias and this alteration is associated with diminished fibrosis and

changes in cardiac miRNA expression.

128

FUTURE DIRECTIONS

The present study focused on investigating the molecular mechanisms underlying

age-related cardiac remodeling in F344XBN rats, with special emphasis on the efficacy

of chronic acetaminophen ingestion as a therapeutic approach. Future studies will be

planned to further extend on the findings of the current study with respect to the

corresponding specific aims as detailed below.

Specific Aim I:

Findings from the present study suggest that aging in male F344XBN rat heart is

associated with increased activation of ERK1/2 and Akt signaling pathways resulting in

increased protein synthesis ultimately leading to cardiac hypertrophy. Future studies will

treat the aged male F344XBN rats with antihypertrophic drugs to test whether a reduction

in cardiac hypertrophy is accompanied by the reduction in the activation of ERK1/2 and

Akt. Results from such studies will no doubt further the molecular signaling profile

underlying age-related cardiac hypertrophy. Whether aging in female F344XBN rat

hearts is associated with similar cardiac remodelling or not remain unexplored. Studies

using young, adult and aged ovariectomized or non-ovariectomized female F344XBN

rats will help to better understand the influence of age and gender on cardiac structure

and function in older age.

Specific Aims II and III

Findings from the present study suggest that chronic acetaminophen ingestion is

able to diminish age-related increases in cardiac oxidative-nitrosative stress, cardiac

129

apoptosis, myocardial fibrosis and the incidence of arrhythmias in male F344XBN rats.

Still, little is known about the mechanisms of cardioprotection employed by chronic

acetaminophen treatment. Previous reports suggest that acetaminophen may act as an

antioxidant as it is a mono-phenol [23]. Future studies will be directed at investigating the

effect of chronic acetaminophen treatment on the expression and activity of antioxidant

enzyme systems in aged male F344XBN rat hearts. Findings from such studies will

provide additional mechanistic data to delineate whether the cardioprotective properties

of acetaminophen are through free radical scavenging or by enhancement of inherent

antioxidant enzymes systems. Moreover, as the majority of studies examining the

cardioprotective properties of acetaminophen were conducted in male rats, the effect of

gender on such cardioprotection conferred by acetaminophen remain largely unknown.

130

REFERENCES

1. Karavidas, A., et al., Aging and the cardiovascular system. Hellenic J Cardiol, 2010. 51(5): p. 421-7.

2. Gerstenblith, G., et al., Echocardiographic assessment of a normal adult aging population. Circulation, 1977. 56(2): p. 273-8.

3. Lakatta, E.G., Age-associated cardiovascular changes in health: impact on cardiovascular disease in older persons. Heart Fail Rev, 2002. 7(1): p. 29-49.

4. Jacobzone S, O.H., Ageing and Health Care Costs. Internationale Politik und Gesellschaft Online (International Politics and Society) 1/2002.

5. Juhaszova, M., et al., Protection in the aged heart: preventing the heart-break of old age? Cardiovasc Res, 2005. 66(2): p. 233-44.

6. Tyagi, P. and G. Tayal, Ischemic preconditioning of myocardium. Acta Pharmacol Sin, 2002. 23(10): p. 865-70.

7. Boluyt, M.O., O.H. Bing, and E.G. Lakatta, The ageing spontaneously hypertensive rat as a model of the transition from stable compensated hypertrophy to heart failure. Eur Heart J, 1995. 16 Suppl N: p. 19-30.

8. d'Alessio, P., Aging and the endothelium. Exp Gerontol, 2004. 39(2): p. 165-71. 9. Delp, M.D., et al., Rat aortic vasoreactivity is altered by old age and hindlimb

unloading. J Appl Physiol, 1995. 78(6): p. 2079-86. 10. Josephson, I.R., et al., Alterations in properties of L-type Ca channels in aging rat

heart. J Mol Cell Cardiol, 2002. 34(3): p. 297-308. 11. Moreau, R., et al., Reversal by aminoguanidine of the age-related increase in

glycoxidation and lipoxidation in the cardiovascular system of Fischer 344 rats. Biochem Pharmacol, 2005. 69(1): p. 29-40.

12. Tschudi, M.R., et al., Effect of age on kinetics of nitric oxide release in rat aorta and pulmonary artery. J Clin Invest, 1996. 98(4): p. 899-905.

13. Lipman, R.D., Pathobiology of aging rodents: inbred and hybrid models. Exp Gerontol, 1997. 32(1-2): p. 215-28.

14. Lipman, R.D., G.E. Dallal, and R.T. Bronson, Effects of genotype and diet on age-related lesions in ad libitum fed and calorie-restricted F344, BN, and BNF3F1 rats. J Gerontol A Biol Sci Med Sci, 1999. 54(11): p. B478-91.

15. Turturro, A., et al., Growth curves and survival characteristics of the animals used in the Biomarkers of Aging Program. J Gerontol A Biol Sci Med Sci, 1999. 54(11): p. B492-501.

16. Hacker, T.A., et al., Age-related changes in cardiac structure and function in Fischer 344 x Brown Norway hybrid rats. Am J Physiol Heart Circ Physiol, 2006. 290(1): p. H304-11.

17. Walker, E.M., Jr., et al., Age-associated changes in hearts of male Fischer 344/Brown Norway F1 rats. Ann Clin Lab Sci, 2006. 36(4): p. 427-38.

131

18. Asano, S., et al., Aging influences multiple indices of oxidative stress in the heart of the Fischer 344/NNia x Brown Norway/BiNia rat. Redox Rep, 2007. 12(4): p. 167-80.

19. Tsutsui, H., S. Kinugawa, and S. Matsushima, Mitochondrial oxidative stress and dysfunction in myocardial remodelling. Cardiovasc Res, 2009. 81(3): p. 449-56.

20. Kakarla, S.K., et al., Possible molecular mechanisms underlying age-related cardiomyocyte apoptosis in the F344XBN rat heart. J Gerontol A Biol Sci Med Sci, 2010. 65(2): p. 147-55.

21. Spiler, N.M., T.H. Rork, and G.F. Merrill, An old drug with a new purpose: cardiovascular actions of acetaminophen (paracetamol). Curr Drug Targets Cardiovasc Haematol Disord, 2005. 5(5): p. 419-29.

22. Ameer, B. and D.J. Greenblatt, Acetaminophen. Ann Intern Med, 1977. 87(2): p. 202-9.

23. Dinis, T.C., V.M. Maderia, and L.M. Almeida, Action of phenolic derivatives (acetaminophen, salicylate, and 5-aminosalicylate) as inhibitors of membrane lipid peroxidation and as peroxyl radical scavengers. Arch Biochem Biophys, 1994. 315(1): p. 161-9.

24. Rork, T.H., et al., Acetaminophen in the hypoxic and reoxygenated guinea pig myocardium. Exp Biol Med (Maywood), 2004. 229(11): p. 1154-61.

25. Merrill, G.F., Acetaminophen and low-flow myocardial ischemia: efficacy and antioxidant mechanisms. Am J Physiol Heart Circ Physiol, 2002. 282(4): p. H1341-9.

26. Golfetti, R., et al., Acetaminophen in the post-ischemia reperfused myocardium. Exp Biol Med (Maywood), 2002. 227(11): p. 1031-7.

27. Merrill, G.F. and E. Goldberg, Antioxidant properties of acetaminophen and cardioprotection. Basic Res Cardiol, 2001. 96(5): p. 423-30.

28. Golfetti, R., T. Rork, and G. Merrill, Chronically administered acetaminophen and the ischemia/reperfused myocardium. Exp Biol Med (Maywood), 2003. 228(6): p. 674-82.

29. Merrill, G., et al., Coronary and myocardial effects of acetaminophen: protection during ischemia-reperfusion. Am J Physiol Heart Circ Physiol, 2001. 280(6): p. H2631-8.

30. Lakatta, E.G., Aging and cardiovascular structure and function in healthy sedentary humans. Aging (Milano), 1998. 10(2): p. 162-4.

31. Harman, D., The free radical theory of aging. Antioxid Redox Signal, 2003. 5(5): p. 557-61.

32. Lakatta, E.G., Cardiovascular Aging: Perspectives From Humans to Rodents. Am J Geriatr Cardiol, 1998. 7(6): p. 32-45.

33. World Population Ageing, 2009; United Nations Department of Economic and Social Affairs/Population Division;ESA/P/WP/212 December 2009.

34. de Grey, A.D., et al., Antiaging technology and pseudoscience. Science, 2002. 296(5568): p. 656.

132

35. World Health Organization. World Health Report 2002, Annex Table 2 (deaths by cause, sex and mortality stratum in WHO Regions, estimates for 2001). Geneva, Switzerland: World Health Organization, 2002:186--91.

36. Kinsella K, V.V., U.S. Census Bureau. An Aging World: 2001. Washington, DC: U.S. Government Printing Office, 2001; series P95/01-1. 2001.

37. Donald G. Fowles, Saadia Greenberg; A Profile of Older Americans: 2009 ; Administration on Aging (AoA), U.S. .

38. Seidell, J.C. and T.L. Visscher, Body weight and weight change and their health implications for the elderly. Eur J Clin Nutr, 2000. 54 Suppl 3: p. S33-9.

39. Short, K.R. and K.S. Nair, Mechanisms of sarcopenia of aging. J Endocrinol Invest, 1999. 22(5 Suppl): p. 95-105.

40. Karakelides, H. and K.S. Nair, Sarcopenia of aging and its metabolic impact. Curr Top Dev Biol, 2005. 68: p. 123-48.

41. Fukagawa, N.K., L.G. Bandini, and J.B. Young, Effect of age on body composition and resting metabolic rate. Am J Physiol, 1990. 259(2 Pt 1): p. E233-8.

42. Harman, D., Aging: a theory based on free radical and radiation chemistry. J Gerontol, 1956. 11(3): p. 298-300.

43. Hulbert, A.J., et al., Life and death: metabolic rate, membrane composition, and life span of animals. Physiol Rev, 2007. 87(4): p. 1175-213.

44. Wahba, W.M., Influence of aging on lung function--clinical significance of changes from age twenty. Anesth Analg, 1983. 62(8): p. 764-76.

45. Leblanc, P., F. Ruff, and J. Milic-Emili, Effects of age and body position on "airway closure" in man. J Appl Physiol, 1970. 28(4): p. 448-51.

46. Martin, J.E. and M.T. Sheaff, Renal ageing. J Pathol, 2007. 211(2): p. 198-205. 47. Domenech, R.J. and P. Macho, [Cardiovascular aging]. Rev Med Chil, 2008.

136(12): p. 1582-8. 48. Cafagna, D. and E. Ponte, [Morphological and functional aspects of the

cardiovascular system related to aging: does "aging heart" exist?]. Minerva Med, 1997. 88(12): p. 491-500.

49. Ferrari, A.U., A. Radaelli, and M. Centola, Invited review: aging and the cardiovascular system. J Appl Physiol, 2003. 95(6): p. 2591-7.

50. Frey, N. and E.N. Olson, Cardiac hypertrophy: the good, the bad, and the ugly. Annu Rev Physiol, 2003. 65: p. 45-79.

51. Downes, T.R., et al., Mechanism of altered pattern of left ventricular filling with aging in subjects without cardiac disease. Am J Cardiol, 1989. 64(8): p. 523-7.

52. Bikkina, M., M.G. Larson, and D. Levy, Prognostic implications of asymptomatic ventricular arrhythmias: the Framingham Heart Study. Ann Intern Med, 1992. 117(12): p. 990-6.

53. Haider, A.W., et al., Increased left ventricular mass and hypertrophy are associated with increased risk for sudden death. J Am Coll Cardiol, 1998. 32(5): p. 1454-9.

133

54. Morales, M.A., et al., Age dependency of myocardial structure: a quantitative two-dimensional echocardiographic study in a normal population. Echocardiography, 2000. 17(3): p. 201-8.

55. Zaragoza, C., et al., Animal models of cardiovascular diseases. J Biomed Biotechnol, 2011. 2011: p. 497841.

56. Anversa, P., et al., Myocyte cell loss and myocyte cellular hyperplasia in the hypertrophied aging rat heart. Circ Res, 1990. 67(4): p. 871-85.

57. Anversa, P. and J.M. Capasso, Cellular basis of aging in the mammalian heart. Scanning Microsc, 1991. 5(4): p. 1065-73; discussion 1073-4.

58. Boluyt, M.O., et al., Echocardiographic assessment of age-associated changes in systolic and diastolic function of the female F344 rat heart. J Appl Physiol, 2004. 96(2): p. 822-8.

59. Sohal, R.S., L.A. Arnold, and B.H. Sohal, Age-related changes in antioxidant enzymes and prooxidant generation in tissues of the rat with special reference to parameters in two insect species. Free Radic Biol Med, 1990. 9(6): p. 495-500.

60. Runge, M.S., K. Molnar, and N.R. Madamanchi, "Old" hearts and arteries: the role of oxidative stress. Trans Am Clin Climatol Assoc, 2010. 121: p. 52-8; discussion 59-60.

61. Anilkumar, N., A. Sirker, and A.M. Shah, Redox sensitive signaling pathways in cardiac remodeling, hypertrophy and failure. Front Biosci, 2009. 14: p. 3168-87.

62. Kinugawa, S., et al., Treatment with dimethylthiourea prevents left ventricular remodeling and failure after experimental myocardial infarction in mice: role of oxidative stress. Circ Res, 2000. 87(5): p. 392-8.

63. Nozik-Grayck, E. and K.R. Stenmark, Role of reactive oxygen species in chronic hypoxia-induced pulmonary hypertension and vascular remodeling. Adv Exp Med Biol, 2007. 618: p. 101-12.

64. Siwik, D.A. and W.S. Colucci, Regulation of matrix metalloproteinases by cytokines and reactive oxygen/nitrogen species in the myocardium. Heart Fail Rev, 2004. 9(1): p. 43-51.

65. de la Asuncion, J.G., et al., Mitochondrial glutathione oxidation correlates with age-associated oxidative damage to mitochondrial DNA. FASEB J, 1996. 10(2): p. 333-8.

66. Shigenaga, M.K., T.M. Hagen, and B.N. Ames, Oxidative damage and mitochondrial decay in aging. Proc Natl Acad Sci U S A, 1994. 91(23): p. 10771-8.

67. Bejma, J., P. Ramires, and L.L. Ji, Free radical generation and oxidative stress with ageing and exercise: differential effects in the myocardium and liver. Acta Physiol Scand, 2000. 169(4): p. 343-51.

68. Kim, J.D., R.J. McCarter, and B.P. Yu, Influence of age, exercise, and dietary restriction on oxidative stress in rats. Aging (Milano), 1996. 8(2): p. 123-9.

69. Nohl, H., V. Breuninger, and D. Hegner, Influence of mitochondrial radical formation on energy-linked respiration. Eur J Biochem, 1978. 90(2): p. 385-90.

134

70. Sohal, R.S. and B.H. Sohal, Hydrogen peroxide release by mitochondria increases during aging. Mech Ageing Dev, 1991. 57(2): p. 187-202.

71. Yu, B.P., Cellular defenses against damage from reactive oxygen species. Physiol Rev, 1994. 74(1): p. 139-62.

72. Kevin, L.G., E. Novalija, and D.F. Stowe, Reactive oxygen species as mediators of cardiac injury and protection: the relevance to anesthesia practice. Anesth Analg, 2005. 101(5): p. 1275-87.

73. Nabeebaccus, A., M. Zhang, and A.M. Shah, NADPH oxidases and cardiac remodelling. Heart Fail Rev, 2011. 16(1): p. 5-12.

74. Cave, A., et al., NADPH oxidase-derived reactive oxygen species in cardiac pathophysiology. Philos Trans R Soc Lond B Biol Sci, 2005. 360(1464): p. 2327-34.

75. Nakamura, K., et al., Inhibitory effects of antioxidants on neonatal rat cardiac myocyte hypertrophy induced by tumor necrosis factor-alpha and angiotensin II. Circulation, 1998. 98(8): p. 794-9.

76. Delbosc, S., et al., Simvastatin prevents angiotensin II-induced cardiac alteration and oxidative stress. Hypertension, 2002. 40(2): p. 142-7.

77. Sabri, A., H.H. Hughie, and P.A. Lucchesi, Regulation of hypertrophic and apoptotic signaling pathways by reactive oxygen species in cardiac myocytes. Antioxid Redox Signal, 2003. 5(6): p. 731-40.

78. Ghosh, M.C., et al., Regulation of calcineurin by oxidative stress. Methods Enzymol, 2003. 366: p. 289-304.

79. Izumiya, Y., et al., Apoptosis signal-regulating kinase 1 plays a pivotal role in angiotensin II-induced cardiac hypertrophy and remodeling. Circ Res, 2003. 93(9): p. 874-83.

80. Kwon, S.H., et al., H(2)O(2) regulates cardiac myocyte phenotype via concentration-dependent activation of distinct kinase pathways. J Mol Cell Cardiol, 2003. 35(6): p. 615-21.

81. von Harsdorf, R., P.F. Li, and R. Dietz, Signaling pathways in reactive oxygen species-induced cardiomyocyte apoptosis. Circulation, 1999. 99(22): p. 2934-41.

82. Flora, S.J., Role of free radicals and antioxidants in health and disease. Cell Mol Biol (Noisy-le-grand), 2007. 53(1): p. 1-2.

83. Nishida, K. and K. Otsu, Cell death in heart failure. Circ J, 2008. 72 Suppl A: p. A17-21.

84. Centurione, L., et al., Age-related death-survival balance in myocardium: an immunohistochemical and biochemical study. Mech Ageing Dev, 2002. 123(4): p. 341-50.

85. Olivetti, G., et al., Cardiomyopathy of the aging human heart. Myocyte loss and reactive cellular hypertrophy. Circ Res, 1991. 68(6): p. 1560-8.

86. Melissari, M., et al., [The aging of the heart: weight and structural changes in the left ventricle with age]. G Ital Cardiol, 1991. 21(2): p. 119-30.

87. Anversa, P., et al., Myocyte cell loss and myocyte hypertrophy in the aging rat heart. J Am Coll Cardiol, 1986. 8(6): p. 1441-8.

135

88. Kajstura, J., et al., Necrotic and apoptotic myocyte cell death in the aging heart of Fischer 344 rats. Am J Physiol, 1996. 271(3 Pt 2): p. H1215-28.

89. Wang, X., et al., The cellular response to oxidative stress: influences of mitogen-activated protein kinase signalling pathways on cell survival. Biochem J, 1998. 333 ( Pt 2): p. 291-300.

90. Xia, Z., et al., Opposing effects of ERK and JNK-p38 MAP kinases on apoptosis. Science, 1995. 270(5240): p. 1326-31.

91. Sugden, P.H. and A. Clerk, "Stress-responsive" mitogen-activated protein kinases (c-Jun N-terminal kinases and p38 mitogen-activated protein kinases) in the myocardium. Circ Res, 1998. 83(4): p. 345-52.

92. Mizukami, Y., et al., Phosphorylation of proteins and apoptosis induced by c-Jun N-terminal kinase1 activation in rat cardiomyocytes by H(2)O(2) stimulation. Biochim Biophys Acta, 2001. 1540(3): p. 213-20.

93. Clerk, A., et al., Stimulation of "stress-regulated" mitogen-activated protein kinases (stress-activated protein kinases/c-Jun N-terminal kinases and p38-mitogen-activated protein kinases) in perfused rat hearts by oxidative and other stresses. J Biol Chem, 1998. 273(13): p. 7228-34.

94. Yue, T.L., et al., Inhibition of extracellular signal-regulated kinase enhances Ischemia/Reoxygenation-induced apoptosis in cultured cardiac myocytes and exaggerates reperfusion injury in isolated perfused heart. Circ Res, 2000. 86(6): p. 692-9.

95. Kumar, D., et al., Apoptosis in adriamycin cardiomyopathy and its modulation by probucol. Antioxid Redox Signal, 2001. 3(1): p. 135-45.

96. Jacobson, M.D., et al., Bcl-2 blocks apoptosis in cells lacking mitochondrial DNA. Nature, 1993. 361(6410): p. 365-9.

97. Cook, S.A., P.H. Sugden, and A. Clerk, Regulation of bcl-2 family proteins during development and in response to oxidative stress in cardiac myocytes: association with changes in mitochondrial membrane potential. Circ Res, 1999. 85(10): p. 940-9.

98. Wolter, K.G., et al., Movement of Bax from the cytosol to mitochondria during apoptosis. J Cell Biol, 1997. 139(5): p. 1281-92.

99. Zha, H., et al., Proapoptotic protein Bax heterodimerizes with Bcl-2 and homodimerizes with Bax via a novel domain (BH3) distinct from BH1 and BH2. J Biol Chem, 1996. 271(13): p. 7440-4.

100. Burlacu, A., Regulation of apoptosis by Bcl-2 family proteins. J Cell Mol Med, 2003. 7(3): p. 249-57.

101. Danial, N.N. and S.J. Korsmeyer, Cell death: critical control points. Cell, 2004. 116(2): p. 205-19.

102. Sharpe, J.C., D. Arnoult, and R.J. Youle, Control of mitochondrial permeability by Bcl-2 family members. Biochim Biophys Acta, 2004. 1644(2-3): p. 107-13.

136

103. Oltvai, Z.N., C.L. Milliman, and S.J. Korsmeyer, Bcl-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programmed cell death. Cell, 1993. 74(4): p. 609-19.

104. Hengartner, M.O., The biochemistry of apoptosis. Nature, 2000. 407(6805): p. 770-6.

105. Phaneuf, S. and C. Leeuwenburgh, Cytochrome c release from mitochondria in the aging heart: a possible mechanism for apoptosis with age. Am J Physiol Regul Integr Comp Physiol, 2002. 282(2): p. R423-30.

106. Song, H.P., et al., The phosphatidylinositol 3-kinase-Akt pathway protects cardiomyocytes from ischaemic and hypoxic apoptosis via mitochondrial function. Clin Exp Pharmacol Physiol, 2010. 37(5-6): p. 598-604.

107. Song, G., G. Ouyang, and S. Bao, The activation of Akt/PKB signaling pathway and cell survival. J Cell Mol Med, 2005. 9(1): p. 59-71.

108. Daneshvar, D., et al., Diastolic dysfunction: improved understanding using emerging imaging techniques. Am Heart J, 2010. 160(3): p. 394-404.

109. Levy, D., et al., Prognostic implications of echocardiographically determined left ventricular mass in the Framingham Heart Study. N Engl J Med, 1990. 322(22): p. 1561-6.

110. Badano, L., et al., [Age-induced changes in the cardiovascular system in normal subjects]. G Ital Cardiol, 1992. 22(9): p. 1023-34.

111. Kitzman, D.W., Diastolic heart failure in the elderly. Heart Fail Rev, 2002. 7(1): p. 17-27.

112. Barry, S.P., S.M. Davidson, and P.A. Townsend, Molecular regulation of cardiac hypertrophy. Int J Biochem Cell Biol, 2008. 40(10): p. 2023-39.

113. Schaub, M.C., et al., Various hypertrophic stimuli induce distinct phenotypes in cardiomyocytes. J Mol Med, 1997. 75(11-12): p. 901-20.

114. Hunter, J.J. and K.R. Chien, Signaling pathways for cardiac hypertrophy and failure. N Engl J Med, 1999. 341(17): p. 1276-83.

115. Stanley, W.C. and M.P. Chandler, Energy metabolism in the normal and failing heart: potential for therapeutic interventions. Heart Fail Rev, 2002. 7(2): p. 115-30.

116. Sugden, P.H., Signalling pathways in cardiac myocyte hypertrophy. Ann Med, 2001. 33(9): p. 611-22.

117. Nicol, R.L., et al., Activated MEK5 induces serial assembly of sarcomeres and eccentric cardiac hypertrophy. Embo J, 2001. 20(11): p. 2757-67.

118. Ikeda, Y., et al., Androgen receptor gene knockout male mice exhibit impaired cardiac growth and exacerbation of angiotensin II-induced cardiac fibrosis. J Biol Chem, 2005. 280(33): p. 29661-6.

119. Sanna, B., et al., Direct and indirect interactions between calcineurin-NFAT and MEK1-extracellular signal-regulated kinase 1/2 signaling pathways regulate cardiac gene expression and cellular growth. Mol Cell Biol, 2005. 25(3): p. 865-78.

137

120. Molkentin, J.D., Calcineurin-NFAT signaling regulates the cardiac hypertrophic response in coordination with the MAPKs. Cardiovasc Res, 2004. 63(3): p. 467-75.

121. McMullen, J.R. and G.L. Jennings, Differences between pathological and physiological cardiac hypertrophy: novel therapeutic strategies to treat heart failure. Clin Exp Pharmacol Physiol, 2007. 34(4): p. 255-62.

122. Heineke, J. and J.D. Molkentin, Regulation of cardiac hypertrophy by intracellular signalling pathways. Nat Rev Mol Cell Biol, 2006. 7(8): p. 589-600.

123. Proud, C.G., Ras, PI3-kinase and mTOR signaling in cardiac hypertrophy. Cardiovasc Res, 2004. 63(3): p. 403-13.

124. Oh, H., et al., Activation of phosphatidylinositol 3-kinase through glycoprotein 130 induces protein kinase B and p70 S6 kinase phosphorylation in cardiac myocytes. J Biol Chem, 1998. 273(16): p. 9703-10.

125. Sugden, P.H. and A. Clerk, Cellular mechanisms of cardiac hypertrophy. J Mol Med, 1998. 76(11): p. 725-46.

126. Aikawa, R., et al., Oxidative stress activates extracellular signal-regulated kinases through Src and Ras in cultured cardiac myocytes of neonatal rats. J Clin Invest, 1997. 100(7): p. 1813-21.

127. Sabri, A., et al., Hydrogen peroxide activates mitogen-activated protein kinases and Na+-H+ exchange in neonatal rat cardiac myocytes. Circ Res, 1998. 82(10): p. 1053-62.

128. Ziolo, M.T., H. Katoh, and D.M. Bers, Expression of inducible nitric oxide synthase depresses beta-adrenergic-stimulated calcium release from the sarcoplasmic reticulum in intact ventricular myocytes. Circulation, 2001. 104(24): p. 2961-6.

129. Proud, C.G., mTORC1 signalling and mRNA translation. Biochem Soc Trans, 2009. 37(Pt 1): p. 227-31.

130. Dorn, G.W., 2nd and T. Force, Protein kinase cascades in the regulation of cardiac hypertrophy. J Clin Invest, 2005. 115(3): p. 527-37.

131. McMullen, J.R., et al., Inhibition of mTOR signaling with rapamycin regresses established cardiac hypertrophy induced by pressure overload. Circulation, 2004. 109(24): p. 3050-5.

132. Bodine, S.C., et al., Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat Cell Biol, 2001. 3(11): p. 1014-9.

133. Gingras, A.C., B. Raught, and N. Sonenberg, Control of translation by the target of rapamycin proteins. Prog Mol Subcell Biol, 2001. 27: p. 143-74.

134. Lakatta, E.G. and D. Levy, Arterial and cardiac aging: major shareholders in cardiovascular disease enterprises: Part II: the aging heart in health: links to heart disease. Circulation, 2003. 107(2): p. 346-54.

135. Kitzman, D.W., et al., Importance of heart failure with preserved systolic function in patients > or = 65 years of age. CHS Research Group. Cardiovascular Health Study. Am J Cardiol, 2001. 87(4): p. 413-9.

138

136. Capasso, J.M., et al., Severe myocardial dysfunction induced by ventricular remodeling in aging rat hearts. Am J Physiol, 1990. 259(4 Pt 2): p. H1086-96.

137. Gielen, S., et al., Aging and heart failure--similar syndromes of exercise intolerance? Implications for exercise-based interventions. Heart Fail Monit, 2005. 4(4): p. 130-6.

138. Stewart, S., et al., Heart failure and the aging population: an increasing burden in the 21st century? Heart, 2003. 89(1): p. 49-53.

139. Creemers, E.E. and Y.M. Pinto, Molecular mechanisms that control interstitial fibrosis in the pressure-overloaded heart. Cardiovasc Res, 2011. 89(2): p. 265-72.

140. Chen, W. and N.G. Frangogiannis, The role of inflammatory and fibrogenic pathways in heart failure associated with aging. Heart Fail Rev, 2010. 15(5): p. 415-22.

141. Centurione, L., et al., Correlations between protein kinase C zeta signaling and morphological modifications during rat heart development and aging. Mech Ageing Dev, 2003. 124(8-9): p. 957-66.

142. Eghbali, M., et al., Collagen accumulation in heart ventricles as a function of growth and aging. Cardiovasc Res, 1989. 23(8): p. 723-9.

143. Orlandi, A., et al., Role of ageing and coronary atherosclerosis in the development of cardiac fibrosis in the rabbit. Cardiovasc Res, 2004. 64(3): p. 544-52.

144. Lin, J., et al., Age-related cardiac muscle sarcopenia: Combining experimental and mathematical modeling to identify mechanisms. Exp Gerontol, 2008. 43(4): p. 296-306.

145. Sawada, M. and J.C. Carlson, Changes in superoxide radical and lipid peroxide formation in the brain, heart and liver during the lifetime of the rat. Mech Ageing Dev, 1987. 41(1-2): p. 125-37.

146. Siwik, D.A., P.J. Pagano, and W.S. Colucci, Oxidative stress regulates collagen synthesis and matrix metalloproteinase activity in cardiac fibroblasts. Am J Physiol Cell Physiol, 2001. 280(1): p. C53-60.

147. Cheng, T.H., et al., Involvement of reactive oxygen species in angiotensin II-induced endothelin-1 gene expression in rat cardiac fibroblasts. J Am Coll Cardiol, 2003. 42(10): p. 1845-54.

148. Frangogiannis, N.G., et al., Critical role of monocyte chemoattractant protein-1/CC chemokine ligand 2 in the pathogenesis of ischemic cardiomyopathy. Circulation, 2007. 115(5): p. 584-92.

149. Dewald, O., et al., CCL2/Monocyte Chemoattractant Protein-1 regulates inflammatory responses critical to healing myocardial infarcts. Circ Res, 2005. 96(8): p. 881-9.

150. Brooks, W.W. and C.H. Conrad, Myocardial fibrosis in transforming growth factor beta(1)heterozygous mice. J Mol Cell Cardiol, 2000. 32(2): p. 187-95.

151. Barcellos-Hoff, M.H. and T.A. Dix, Redox-mediated activation of latent transforming growth factor-beta 1. Mol Endocrinol, 1996. 10(9): p. 1077-83.

139

152. Park, S.K., et al., Hydrogen peroxide is a novel inducer of connective tissue growth factor. Biochem Biophys Res Commun, 2001. 284(4): p. 966-71.

153. Lee, A.A., et al., Angiotensin II stimulates the autocrine production of transforming growth factor-beta 1 in adult rat cardiac fibroblasts. J Mol Cell Cardiol, 1995. 27(10): p. 2347-57.

154. Campbell, S.E. and L.C. Katwa, Angiotensin II stimulated expression of transforming growth factor-beta1 in cardiac fibroblasts and myofibroblasts. J Mol Cell Cardiol, 1997. 29(7): p. 1947-58.

155. Rosenkranz, S., TGF-beta1 and angiotensin networking in cardiac remodeling. Cardiovasc Res, 2004. 63(3): p. 423-32.

156. Ambros, V., microRNAs: tiny regulators with great potential. Cell, 2001. 107(7): p. 823-6.

157. Lee, R.C., R.L. Feinbaum, and V. Ambros, The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell, 1993. 75(5): p. 843-54.

158. Esquela-Kerscher, A. and F.J. Slack, Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer, 2006. 6(4): p. 259-69.

159. Costinean, S., et al., Pre-B cell proliferation and lymphoblastic leukemia/high-grade lymphoma in E(mu)-miR155 transgenic mice. Proc Natl Acad Sci U S A, 2006. 103(18): p. 7024-9.

160. Hammond, S.M., MicroRNAs as oncogenes. Curr Opin Genet Dev, 2006. 16(1): p. 4-9.

161. Ambros, V., MicroRNA pathways in flies and worms: growth, death, fat, stress, and timing. Cell, 2003. 113(6): p. 673-6.

162. van Rooij, E., et al., A signature pattern of stress-responsive microRNAs that can evoke cardiac hypertrophy and heart failure. Proc Natl Acad Sci U S A, 2006. 103(48): p. 18255-60.

163. Latronico, M.V., D. Catalucci, and G. Condorelli, MicroRNA and cardiac pathologies. Physiol Genomics, 2008. 34(3): p. 239-42.

164. Care, A., et al., MicroRNA-133 controls cardiac hypertrophy. Nat Med, 2007. 13(5): p. 613-8.

165. van Rooij, E., et al., Control of stress-dependent cardiac growth and gene expression by a microRNA. Science, 2007. 316(5824): p. 575-9.

166. Yang, B., et al., The muscle-specific microRNA miR-1 regulates cardiac arrhythmogenic potential by targeting GJA1 and KCNJ2. Nat Med, 2007. 13(4): p. 486-91.

167. Zhao, Y., et al., Dysregulation of cardiogenesis, cardiac conduction, and cell cycle in mice lacking miRNA-1-2. Cell, 2007. 129(2): p. 303-17.

168. Zhao, Y., E. Samal, and D. Srivastava, Serum response factor regulates a muscle-specific microRNA that targets Hand2 during cardiogenesis. Nature, 2005. 436(7048): p. 214-20.

140

169. Ikeda, S., et al., Altered microRNA expression in human heart disease. Physiol Genomics, 2007. 31(3): p. 367-73.

170. Tatsuguchi, M., et al., Expression of microRNAs is dynamically regulated during cardiomyocyte hypertrophy. J Mol Cell Cardiol, 2007. 42(6): p. 1137-41.

171. Shan, Z.X., et al., Upregulated expression of miR-1/miR-206 in a rat model of myocardial infarction. Biochem Biophys Res Commun, 2009. 381(4): p. 597-601.

172. Xu, C., et al., The muscle-specific microRNAs miR-1 and miR-133 produce opposing effects on apoptosis by targeting HSP60, HSP70 and caspase-9 in cardiomyocytes. J Cell Sci, 2007. 120(Pt 17): p. 3045-52.

173. Xiao, J., et al., MicroRNA miR-133 represses HERG K+ channel expression contributing to QT prolongation in diabetic hearts. J Biol Chem, 2007. 282(17): p. 12363-7.

174. Thum, T., et al., MicroRNA-21 contributes to myocardial disease by stimulating MAP kinase signalling in fibroblasts. Nature, 2008. 456(7224): p. 980-4.

175. Cheng, Y., et al., MicroRNAs are aberrantly expressed in hypertrophic heart: do they play a role in cardiac hypertrophy? Am J Pathol, 2007. 170(6): p. 1831-40.

176. Duisters, R.F., et al., miR-133 and miR-30 regulate connective tissue growth factor: implications for a role of microRNAs in myocardial matrix remodeling. Circ Res, 2009. 104(2): p. 170-8, 6p following 178.

177. Lukiw, W.J. and A.I. Pogue, Induction of specific micro RNA (miRNA) species by ROS-generating metal sulfates in primary human brain cells. J Inorg Biochem, 2007. 101(9): p. 1265-9.

178. Shilo, S., et al., Evidence for the involvement of miRNA in redox regulated angiogenic response of human microvascular endothelial cells. Arterioscler Thromb Vasc Biol, 2008. 28(3): p. 471-7.

179. Prescott, L.F., Paracetamol: past, present, and future. Am J Ther, 2000. 7(2): p. 143-7.

180. Spooner, J.B. and J.G. Harvey, The history and usage of paracetamol. J Int Med Res, 1976. 4(4 Suppl): p. 1-6.

181. Anderson, B.J., Paracetamol (Acetaminophen): mechanisms of action. Paediatr Anaesth, 2008. 18(10): p. 915-21.

182. Botting, R.M., Mechanism of action of acetaminophen: is there a cyclooxygenase 3? Clin Infect Dis, 2000. 31 Suppl 5: p. S202-10.

183. Dahlin, D.C., et al., N-acetyl-p-benzoquinone imine: a cytochrome P-450-mediated oxidation product of acetaminophen. Proc Natl Acad Sci U S A, 1984. 81(5): p. 1327-31.

184. James, L.P., P.R. Mayeux, and J.A. Hinson, Acetaminophen-induced hepatotoxicity. Drug Metab Dispos, 2003. 31(12): p. 1499-506.

185. Bertolini, A., et al., Paracetamol: new vistas of an old drug. CNS Drug Rev, 2006. 12(3-4): p. 250-75.

186. Stirnimann, G., K. Kessebohm, and B. Lauterburg, Liver injury caused by drugs: an update. Swiss Med Wkly, 2010. 140: p. w13080.

141

187. Hinson, J.A., D.W. Roberts, and L.P. James, Mechanisms of acetaminophen-induced liver necrosis. Handb Exp Pharmacol, 2010(196): p. 369-405.

188. Tang, W., Drug metabolite profiling and elucidation of drug-induced hepatotoxicity. Expert Opin Drug Metab Toxicol, 2007. 3(3): p. 407-20.

189. Boutaud, O., et al., Determinants of the cellular specificity of acetaminophen as an inhibitor of prostaglandin H(2) synthases. Proc Natl Acad Sci U S A, 2002. 99(10): p. 7130-5.

190. Brennan, M.L., et al., Prognostic value of myeloperoxidase in patients with chest pain. N Engl J Med, 2003. 349(17): p. 1595-604.

191. Chou, T.M. and P. Greenspan, Effect of acetaminophen on the myeloperoxidase-hydrogen peroxide-nitrite mediated oxidation of LDL. Biochim Biophys Acta, 2002. 1581(1-2): p. 57-63.

192. Maharaj, D.S., et al., Acetaminophen and aspirin inhibit superoxide anion generation and lipid peroxidation, and protect against 1-methyl-4-phenyl pyridinium-induced dopaminergic neurotoxicity in rats. Neurochem Int, 2004. 44(5): p. 355-60.

193. Nakamoto, K., et al., Protective effect of acetaminophen against acute gastric mucosal lesions induced by ischemia-reperfusion in the rat. Pharmacology, 1997. 54(4): p. 203-10.

194. Cheung, P.Y., et al., Matrix metalloproteinase-2 contributes to ischemia-reperfusion injury in the heart. Circulation, 2000. 101(15): p. 1833-9.

195. Wang, W., G. Sawicki, and R. Schulz, Peroxynitrite-induced myocardial injury is mediated through matrix metalloproteinase-2. Cardiovasc Res, 2002. 53(1): p. 165-74.

196. Rork, T.H., et al., Acetaminophen attenuates peroxynitrite-activated matrix metalloproteinase-2-mediated troponin I cleavage in the isolated guinea pig myocardium. J Mol Cell Cardiol, 2006. 40(4): p. 553-61.

197. Merrill, G.F., et al., Acetaminophen and myocardial infarction in dogs. Am J Physiol Heart Circ Physiol, 2004. 287(5): p. H1913-20.

198. Merrill, G.F., et al., Antiarrhythmic properties of acetaminophen in the dog. Exp Biol Med (Maywood), 2007. 232(9): p. 1245-52.

199. Hadzimichalis, N.M., et al., Acetaminophen-mediated cardioprotection via inhibition of the mitochondrial permeability transition pore-induced apoptotic pathway. Am J Physiol Heart Circ Physiol, 2007. 293(6): p. H3348-55.

200. Jaques-Robinson, K.M., et al., Acetaminophen is cardioprotective against H2O2-induced injury in vivo. Exp Biol Med (Maywood), 2008. 233(10): p. 1315-22.

201. Lakatta, E.G. and S.J. Sollott, The "heartbreak" of older age. Mol Interv, 2002. 2(7): p. 431-46.

202. Lipman, R.D., et al., Pathologic characterization of brown Norway, brown Norway x Fischer 344, and Fischer 344 x brown Norway rats with relation to age. J Gerontol A Biol Sci Med Sci, 1996. 51(1): p. B54-9.

142

203. Kakarla, S.K., et al., Chronic acetaminophen attenuates age-associated increases in cardiac ROS and apoptosis in the Fischer Brown Norway rat. Basic Res Cardiol, 2010. 105(4): p. 535-44.

204. Hannan, R.D., et al., Cardiac hypertrophy: a matter of translation. Clin Exp Pharmacol Physiol, 2003. 30(8): p. 517-27.

205. Aoki, Y., et al., The RAS/MAPK syndromes: novel roles of the RAS pathway in human genetic disorders. Hum Mutat, 2008. 29(8): p. 992-1006.

206. Ueyama, T., et al., Requirement of activation of the extracellular signal-regulated kinase cascade in myocardial cell hypertrophy. J Mol Cell Cardiol, 2000. 32(6): p. 947-60.

207. Yamazaki, T., et al., Mechanical loading activates mitogen-activated protein kinase and S6 peptide kinase in cultured rat cardiac myocytes. J Biol Chem, 1993. 268(16): p. 12069-76.

208. Bogoyevitch, M.A., et al., Endothelin-1 and fibroblast growth factors stimulate the mitogen-activated protein kinase signaling cascade in cardiac myocytes. The potential role of the cascade in the integration of two signaling pathways leading to myocyte hypertrophy. J Biol Chem, 1994. 269(2): p. 1110-9.

209. Bueno, O.F. and J.D. Molkentin, Involvement of extracellular signal-regulated kinases 1/2 in cardiac hypertrophy and cell death. Circ Res, 2002. 91(9): p. 776-81.

210. Yu, W., et al., Insulin signaling: a possible pathogenesis of cardiac hypertrophy. Cardiovasc Ther, 2010. 28(2): p. 101-5.

211. Towbin, H., T. Staehelin, and J. Gordon, Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proc Natl Acad Sci U S A, 1979. 76(9): p. 4350-4.

212. Gupte, A.A., G.L. Bomhoff, and P.C. Geiger, Age-related differences in skeletal muscle insulin signaling: the role of stress kinases and heat shock proteins. J Appl Physiol, 2008. 105(3): p. 839-48.

213. Marzetti, E., et al., Age-related activation of mitochondrial caspase-independent apoptotic signaling in rat gastrocnemius muscle. Mech Ageing Dev, 2008. 129(9): p. 542-9.

214. Wheeler, M.T., S. Zarnegar, and E.M. McNally, Zeta-sarcoglycan, a novel component of the sarcoglycan complex, is reduced in muscular dystrophy. Hum Mol Genet, 2002. 11(18): p. 2147-54.

215. Inoki, K., et al., Signaling by target of rapamycin proteins in cell growth control. Microbiol Mol Biol Rev, 2005. 69(1): p. 79-100.

216. Gingras, A.C., et al., Hierarchical phosphorylation of the translation inhibitor 4E-BP1. Genes Dev, 2001. 15(21): p. 2852-64.

217. Ruvinsky, I., et al., Ribosomal protein S6 phosphorylation is a determinant of cell size and glucose homeostasis. Genes Dev, 2005. 19(18): p. 2199-211.

143

218. Bogoyevitch, M.A., C.J. Marshall, and P.H. Sugden, Hypertrophic agonists stimulate the activities of the protein kinases c-Raf and A-Raf in cultured ventricular myocytes. J Biol Chem, 1995. 270(44): p. 26303-10.

219. Clerk, A., et al., Differential activation of protein kinase C isoforms by endothelin-1 and phenylephrine and subsequent stimulation of p42 and p44 mitogen-activated protein kinases in ventricular myocytes cultured from neonatal rat hearts. J Biol Chem, 1994. 269(52): p. 32848-57.

220. Schluter, K.D., et al., Role of phosphatidylinositol 3-kinase activation in the hypertrophic growth of adult ventricular cardiomyocytes. Cardiovasc Res, 1998. 40(1): p. 174-81.

221. Shioi, T., et al., Akt/protein kinase B promotes organ growth in transgenic mice. Mol Cell Biol, 2002. 22(8): p. 2799-809.

222. Matsui, T., et al., Phenotypic spectrum caused by transgenic overexpression of activated Akt in the heart. J Biol Chem, 2002. 277(25): p. 22896-901.

223. Cantley, L.C., The phosphoinositide 3-kinase pathway. Science, 2002. 296(5573): p. 1655-7.

224. Antos, C.L., et al., Activated glycogen synthase-3 beta suppresses cardiac hypertrophy in vivo. Proc Natl Acad Sci U S A, 2002. 99(2): p. 907-12.

225. Oudit, G.Y. and J.M. Penninger, Cardiac regulation by phosphoinositide 3-kinases and PTEN. Cardiovasc Res, 2009. 82(2): p. 250-60.

226. Wang, X. and C.G. Proud, The mTOR pathway in the control of protein synthesis. Physiology (Bethesda), 2006. 21: p. 362-9.

227. Manning, B.D., et al., Identification of the tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target of the phosphoinositide 3-kinase/akt pathway. Mol Cell, 2002. 10(1): p. 151-62.

228. Wang, L., I. Gout, and C.G. Proud, Cross-talk between the ERK and p70 S6 kinase (S6K) signaling pathways. MEK-dependent activation of S6K2 in cardiomyocytes. J Biol Chem, 2001. 276(35): p. 32670-7.

229. Wang, L. and C.G. Proud, Ras/Erk signaling is essential for activation of protein synthesis by Gq protein-coupled receptor agonists in adult cardiomyocytes. Circ Res, 2002. 91(9): p. 821-9.

230. Nojima, H., et al., The mammalian target of rapamycin (mTOR) partner, raptor, binds the mTOR substrates p70 S6 kinase and 4E-BP1 through their TOR signaling (TOS) motif. J Biol Chem, 2003. 278(18): p. 15461-4.

231. Franklin, S.S., Cardiovascular risks related to increased diastolic, systolic and pulse pressure. An epidemiologist's point of view. Pathol Biol (Paris), 1999. 47(6): p. 594-603.

232. Lakatta, E., Aging Effects on the Vasculature in Health: Risk Factors for Cardiovascular Disease. Am J Geriatr Cardiol, 1994. 3(6): p. 11-17.

233. Lauer, T., et al., Age-dependent endothelial dysfunction is associated with failure to increase plasma nitrite in response to exercise. Basic Res Cardiol, 2008. 103(3): p. 291-7.

144

234. Laurent, S., Arterial wall hypertrophy and stiffness in essential hypertensive patients. Hypertension, 1995. 26(2): p. 355-62.

235. Hare, J.M., Oxidative stress and apoptosis in heart failure progression. Circ Res, 2001. 89(3): p. 198-200.

236. Wencker, D., et al., A mechanistic role for cardiac myocyte apoptosis in heart failure. J Clin Invest, 2003. 111(10): p. 1497-504.

237. Lee, H.C. and Y.H. Wei, Oxidative stress, mitochondrial DNA mutation, and apoptosis in aging. Exp Biol Med (Maywood), 2007. 232(5): p. 592-606.

238. Wu, M., et al., Acetaminophen prevents aging-associated hyperglycemia in aged rats: effect of aging-associated hyperactivation of p38-MAPK and ERK1/2. Diabetes Metab Res Rev, 2009. 25(3): p. 279-86.

239. Zhao, H., et al., Superoxide reacts with hydroethidine but forms a fluorescent product that is distinctly different from ethidium: potential implications in intracellular fluorescence detection of superoxide. Free Radic Biol Med, 2003. 34(11): p. 1359-68.

240. Carter, W.O., P.K. Narayanan, and J.P. Robinson, Intracellular hydrogen peroxide and superoxide anion detection in endothelial cells. J Leukoc Biol, 1994. 55(2): p. 253-8.

241. Comporti, M., Lipid peroxidation and biogenic aldehydes: from the identification of 4-hydroxynonenal to further achievements in biopathology. Free Radic Res, 1998. 28(6): p. 623-35.

242. Los, M., et al., Switching Akt: from survival signaling to deadly response. Bioessays, 2009. 31(5): p. 492-5.

243. Rosner, M., et al., Functional interaction of mammalian target of rapamycin complexes in regulating mammalian cell size and cell cycle. Hum Mol Genet, 2009. 18(17): p. 3298-310.

244. Dhingra, S., et al., p38 and ERK1/2 MAPKs mediate the interplay of TNF-alpha and IL-10 in regulating oxidative stress and cardiac myocyte apoptosis. Am J Physiol Heart Circ Physiol, 2007. 293(6): p. H3524-31.

245. Das, A., et al., ERK phosphorylation mediates sildenafil-induced myocardial protection against ischemia-reperfusion injury in mice. Am J Physiol Heart Circ Physiol, 2009. 296(5): p. H1236-43.

246. Casas-Terradellas, E., et al., ERK and p38 pathways regulate amino acid signalling. Biochim Biophys Acta, 2008. 1783(12): p. 2241-54.

247. Wagner, C., et al., Ischemic post-conditioning reduces infarct size of the in vivo rat heart: role of PI3-K, mTOR, GSK-3beta, and apoptosis. Mol Cell Biochem, 2010.

248. Ungvari, Z., et al., Role of oxidative-nitrosative stress and downstream pathways in various forms of cardiomyopathy and heart failure. Curr Vasc Pharmacol, 2005. 3(3): p. 221-9.

145

249. Dost, T., M.V. Cohen, and J.M. Downey, Redox signaling triggers protection during the reperfusion rather than the ischemic phase of preconditioning. Basic Res Cardiol, 2008. 103(4): p. 378-84.

250. Besse, S., et al., Antioxidant treatment prevents cardiac protein oxidation after ischemia-reperfusion and improves myocardial function and coronary perfusion in senescent hearts. J Physiol Pharmacol, 2006. 57(4): p. 541-52.

251. Berger, J.S., et al., Aspirin for the primary prevention of cardiovascular events in women and men: a sex-specific meta-analysis of randomized controlled trials. Jama, 2006. 295(3): p. 306-13.

252. Ray, S.D., et al., Oxidative stress is the master operator of drug and chemically-induced programmed and unprogrammed cell death: Implications of natural antioxidants in vivo. Biofactors, 2004. 21(1-4): p. 223-32.

253. Boengler, K., R. Schulz, and G. Heusch, Loss of cardioprotection with ageing. Cardiovasc Res, 2009. 83(2): p. 247-61.

254. Griendling, K.K., D. Sorescu, and M. Ushio-Fukai, NAD(P)H oxidase: role in cardiovascular biology and disease. Circ Res, 2000. 86(5): p. 494-501.

255. Adler, A., et al., NAD(P)H oxidase-generated superoxide anion accounts for reduced control of myocardial O2 consumption by NO in old Fischer 344 rats. Am J Physiol Heart Circ Physiol, 2003. 285(3): p. H1015-22.

256. Pletjushkina, O.Y., et al., Hydrogen peroxide produced inside mitochondria takes part in cell-to-cell transmission of apoptotic signal. Biochemistry (Mosc), 2006. 71(1): p. 60-7.

257. Kwak, H.B., W. Song, and J.M. Lawler, Exercise training attenuates age-induced elevation in Bax/Bcl-2 ratio, apoptosis, and remodeling in the rat heart. Faseb J, 2006. 20(6): p. 791-3.

258. Fischer, P. and D. Hilfiker-Kleiner, Survival pathways in hypertrophy and heart failure: the gp130-STAT axis. Basic Res Cardiol, 2007. 102(5): p. 393-411.

259. Balasubramanian, S., et al., mTOR in growth and protection of hypertrophying myocardium. Cardiovasc Hematol Agents Med Chem, 2009. 7(1): p. 52-63.

260. Rosner, M., et al., Functional interaction of mTOR complexes in regulating mammalian cell size and cell cycle. Hum Mol Genet, 2009.

261. Qin, S. and P.B. Chock, Implication of phosphatidylinositol 3-kinase membrane recruitment in hydrogen peroxide-induced activation of PI3K and Akt. Biochemistry, 2003. 42(10): p. 2995-3003.

262. Konishi, H., et al., Activation of protein kinase B (Akt/RAC-protein kinase) by cellular stress and its association with heat shock protein Hsp27. FEBS Lett, 1997. 410(2-3): p. 493-8.

263. Liu, L., et al., Heat shock protein 27 regulates oxidative stress-induced apoptosis in cardiomyocytes: mechanisms via reactive oxygen species generation and Akt activation. Chin Med J (Engl), 2007. 120(24): p. 2271-7.

264. Shimizu, N., et al., Activation of mitogen-activated protein kinases and activator protein-1 in myocardial infarction in rats. Cardiovasc Res, 1998. 38(1): p. 116-24.

146

265. Xu, Z., X. Ji, and P.G. Boysen, Exogenous nitric oxide generates ROS and induces cardioprotection: involvement of PKG, mitochondrial KATP channels, and ERK. Am J Physiol Heart Circ Physiol, 2004. 286(4): p. H1433-40.

266. Li, D.Y., et al., Role of ERK1/2 in the anti-apoptotic and cardioprotective effects of nitric oxide after myocardial ischemia and reperfusion. Apoptosis, 2006. 11(6): p. 923-30.

267. Huikuri, H.V., A. Castellanos, and R.J. Myerburg, Sudden death due to cardiac arrhythmias. N Engl J Med, 2001. 345(20): p. 1473-82.

268. Rice, K.M., et al., Fluprostenol-induced MAPK signaling is independent of aging in Fischer 344/NNiaHSd x Brown Norway/BiNia rat aorta. Ann Clin Lab Sci, 2010. 40(1): p. 26-31.

269. Ide, T., et al., Mitochondrial electron transport complex I is a potential source of oxygen free radicals in the failing myocardium. Circ Res, 1999. 85(4): p. 357-63.

270. Saavedra, W.F., et al., Imbalance between xanthine oxidase and nitric oxide synthase signaling pathways underlies mechanoenergetic uncoupling in the failing heart. Circ Res, 2002. 90(3): p. 297-304.

271. Li, J.M., et al., Activation of NADPH oxidase during progression of cardiac hypertrophy to failure. Hypertension, 2002. 40(4): p. 477-84.

272. Landmesser, U., K.C. Wollert, and H. Drexler, Potential novel pharmacological therapies for myocardial remodelling. Cardiovasc Res, 2009. 81(3): p. 519-27.

273. Rice, K.M., et al., Effects of aging on pressure-induced MAPK activation in the rat aorta. Pflugers Arch, 2005. 450(3): p. 192-9.

274. Marasa, B.S., et al., MicroRNA profiling in human diploid fibroblasts uncovers miR-519 role in replicative senescence. Aging (Albany NY), 2010. 2(6): p. 333-43.

275. Livak, K.J. and T.D. Schmittgen, Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods, 2001. 25(4): p. 402-8.

276. Sangaralingham, S.J., et al., The aging heart, myocardial fibrosis, and its relationship to circulating C-type natriuretic Peptide. Hypertension, 2011. 57(2): p. 201-7.

277. Sasano, C., et al., Internalization and dephosphorylation of connexin43 in hypertrophied right ventricles of rats with pulmonary hypertension. Circ J, 2007. 71(3): p. 382-9.

278. Melov, S., Animal models of oxidative stress, aging, and therapeutic antioxidant interventions. Int J Biochem Cell Biol, 2002. 34(11): p. 1395-400.

279. Danik, S.B., et al., Modulation of cardiac gap junction expression and arrhythmic susceptibility. Circ Res, 2004. 95(10): p. 1035-41.

280. Weber, K.T., C.G. Brilla, and J.S. Janicki, Myocardial fibrosis: functional significance and regulatory factors. Cardiovasc Res, 1993. 27(3): p. 341-8.

281. Sayed, D., et al., MicroRNAs play an essential role in the development of cardiac hypertrophy. Circ Res, 2007. 100(3): p. 416-24.

147

282. Lin, Y., et al., Involvement of MicroRNAs in hydrogen peroxide-mediated gene regulation and cellular injury response in vascular smooth muscle cells. J Biol Chem, 2009. 284(12): p. 7903-13.

283. Cheng, Y., et al., MicroRNA-21 protects against the H(2)O(2)-induced injury on cardiac myocytes via its target gene PDCD4. J Mol Cell Cardiol, 2009. 47(1): p. 5-14.

284. Xiao, J., et al., MicroRNA miR-133 represses HERG K+ channel expression contributing to QT prolongation in diabetic hearts. J Biol Chem, 2007. 282(17): p. 12363-7.

285. Liu, N., et al., microRNA-133a regulates cardiomyocyte proliferation and suppresses smooth muscle gene expression in the heart. Genes Dev, 2008. 22(23): p. 3242-54.

148

CURRICULUM VITAE

SUNIL K. KAKARLA

6633 Country club drive, Huntington, WV 25705

Phone: 919-656-6414. E-mail: [email protected]

OBJECTIVE

To work as an assistant professor in a renowned College of Veterinary Medicine and to

be placed in the elite group of researchers where I can explore the field of science to

quench my thirst for knowledge and grow along with the institute.

EDUCATION

July 2007-Present Ph. D., Biomedical Science (Doctoral candidate) at John C Edward

School of Medicine, Marshall University, Huntington, WV

Dissertation: “AGE-ASSOCIATED ALTERATIONS IN THE

F344XBN RAT HEART AND THE EFFICACY OF

CHRONIC ACETAMINOPHEN INGESTION AS A

THERAPEUTIC APPROACH”

Aug 2005-Jun 2007 MS in Biological Sciences (emphasis in Molecular Physiology) at

Marshall University, Huntington, WV

Thesis: “AGE-RELATED CHANGES IN CARDIAC

DYSTROPHIN GLYCOPROTEIN COMPLEX STRUCTURE

AND FUNCTION IN THE F344XBN RATS”

Nov 1994-Oct 1999 DVM at College of Veterinary Science, ANGRAU, Tirupati, A.P,

INDIA

LICENSURE NC state veterinary license

CERTIFICATIONS Completed Education Commission for Foreign Veterinary

Graduates (ECFVG) certification

PROFESSIONAL EXPERIENCE

RESEARCH EXPERIENCE

149

Aug 2005-Present Research Assistant, Laboratory of Molecular physiology,

Marshall University, Huntington, WV.

July 2002-July 2005 Veterinary Assistant Surgeon, Animal Husbandry

Department, INDIA.

Nov 1999-June 2002 Poultry Pathologist, Vijayanagaram Hatcheries Pvt Ltd,

VSP, A.P, INDIA.

PUBLICATIONS

PUBLISHED

Wang Y, Wu M, Al-Rousan R, Liu H, Fannin J, Paturi S, Arvapalli R, Katta A, Kakarla

SK, Rice K, Triest WE, Blough ER. Iron-induced cardiac damage: role of apoptosis and

deferasirox intervention. J Pharmacol Exp Ther. 2011 Jan;336(1):56-63

Katta A, Kundla S, Kakarla SK, Wu M, Fannin J, Paturi S, Liu H, Addagarla HS,

Blough ER. Impaired overload-induced hypertrophy is associated with diminished mTOR

signaling in insulin resistant skeletal muscle of the obese Zucker rat. Am J Physiol Regul

Integr Comp Physiol. 2010 Dec;299(6):R1666-75.

Wu M, Liu H, Fannin J, Katta A, Wang Y, Arvapalli RK, Paturi S, Kakarla SK, Rice

KM, Blough ER. Acetaminophen Improves Protein Translational Signaling in Aged

Skeletal Muscle. Rejuvenation Res. 2010 Oct;13(5):571-9

Kakarla SK, Fannin JC, Keshavarzian S, Katta A, Paturi S, Nalabotu SK, Wu M, Rice

KM, Manzoor K, Walker EM Jr, Blough ER; Chronic acetaminophen attenuates age-

associated increases in cardiac ROS and apoptosis in the Fischer Brown Norway Hybrid

Rat. Basic Res Cardiol. 2010 Jul; 105(4):535-44

Kolli MB, Day BS, Takatsuki H, Nalabotu SK, Rice KM, Kohama K, Gadde MK,

Kakarla SK, Katta A, Blough ER; Application of PAMAM dendrimers for use in

bionanomotor systems. Langmuir. 2010 May 4; 26(9):6079-82

Arvapalli RK, Paturi S, Laurino JP, Katta A, Kakarla SK, Gadde MK, Wu M, Rice KM,

Walker EM, Wehner P, Blough ER. Deferasirox decreases age-associated iron

accumulation in the aging F344XBN rat heart. Cardiovasc Toxicol. 2010 Jun; 10(2):108-

16

150

Paturi S, Gutta AK, Katta A, Kakarla SK, Arvapalli RK, Gadde MK, Nalabotu SK, Rice

KM, Wu M, Blough E; Effects of aging and gender on regulators of muscle adaptation in

the aging F344/BN rat model. Mech Ageing Dev. 2010 Mar; 131(3):202-9

Rice KM, Walker EM, Kakarla SK, Paturi S, Wu M, Narula S, Blough ER; Aging Does

Not Alters Fluprostenol-Induced MAPK Signaling in the Fischer 344/NNiaHSd X Brown

Norway /BiNia Rat Aorta. Ann Clin Lab Sci. 2010 Winter; 40 (1):26-31

Kakarla SK, Rice KM, Katta A, Paturi S, Wu M, Kolli M, Keshavarzian S, Manzoor K,

Wehner PS, Blough ER; Possible molecular mechanisms underlying age-related

cardiomyocyte apoptosis in the F344XBN Rat Heart. J Gerontol A Biol Sci Med Sci.

2010 Feb; 65(2):147-55.

Kakarla SK, Rice KM, Mupparaju SP, Paturi S, Katta A, Wu M, Harris RT, Blough ER;

Shear stress activates Akt during vascular smooth muscle cell reorientation. Biotechnol

Appl Biochem. 2010 Feb 18; 55(2):85-90.

Al-Rousan RM, Paturi S, Laurino JP, Kakarla SK, Gutta AK, Walker EM, Blough ER.

2009. Deferasirox Removes Cardiac Iron and Attenuates Oxidative Stress in the Iron-

Overloaded Gerbil. Am J Hematol. 2009 Sep; 84(9):565-70.

Paturi S, Gutta AK, Kakarla SK, Katta A, Arnold EC, Wu M, Rice KM, Blough ER;

Impaired overload-induced hypertrophy in obese zucker rat slow-twitch skeletal muscle.

J Appl Physiol. 2010 Jan; 108(1):7-13. Epub 2009 Sep 24.

Wu M, Katta A, Gadde MK, Liu H, Kakarla SK, Fannin J, Paturi S, Arvapalli RK, Rice

KM, Wang Y, Blough ER; Aging-associated dysfunction of Akt / protein kinase B: S-

nitrosylation and acetaminophen intervention. PLoS One. 2009 Jul 29; 4(7):e6430.

Wu M, Desai DH, Kakarla SK, Katta A, Paturi S, Gutta AK, Rice KM, Walker EM Jr,

Blough ER. Acetaminophen prevents aging-associated hyperglycemia in aged rats: effect

of aging-associated hyperactivation of p38-MAPK and ERK1/2. Diabetes Metab Res

Rev. 2009 Mar; 25(3):279-86.

Katta A, Kakarla SK, Wu M, Meduru S, Desai DH, Rice KM, Blough ER. Lean and

obese Zucker rats exhibit different patterns of p70s6 kinase regulation in the tibialis

anterior muscle in response to high-force muscle contraction. Muscle Nerve. 2009 Apr;

39(4):503-11.

151

Kakarla SK, Katta A, Wu M, Paturi S, Gadde MK, Arvapalli R, Kolli M, Rice KM,

Blough ER; Altered regulation of contraction-induced Akt / mTOR / p70S6k pathway

signaling in skeletal muscle of Syndrome-X Obese Zucker Rat Model. Exp Diabetes Res.

2009; 2009:384683.

Katta A, Preston DL, Kakarla SK, Asano S, Meduru S, Mupparaju SP, Yokochi E, Rice

KM, Desai DH, Blough ER. Diabetes alters contraction-induced mitogen activated

protein kinase activation in the rat soleus and plantaris. Exp Diabetes Res. 2008;

2008:738101.

Asano S, Rice KM, Kakarla SK, Katta A, Desai DH, Walker EM, Wehner P, Blough

ER. Aging influences multiple indices of oxidative stress in the heart of the Fischer

344/NNia x Brown Norway/BiNia rat. Redox Rep. 2007; 12(4):167-80.

Rice KM, Desai DH, Kakarla SK, Katta A, Preston DL, Wehner P, Blough ER. Diabetes

alters vascular mechanotransduction: pressure-induced regulation of mitogen activated

protein kinases in the rat inferior vena cava. Cardiovasc Diabetol. 2006 Sep 8; 5:18.

MANUSCRIPTS IN REVIEW

Kakarla SK, Thulluri S, Manne N.D.P.K, Rice KM, Blough ER; Anti-arrhythmic effect

of chronic acetaminophen treatment in the aging F344XBN rat involves diminished

myocardial fibrosis and altered miRNAs regulation.

Kakarla SK, Sudarsanam K, Kolli MB, Katta A, Nalabotu SK, Rice KM, Blough ER;

Mechanisms of age-related cardiac hypertrophy in the F344XBN rat model.

Anjaiah K, kakarla SK, Wu M, Nalabotu SK, Rice KM, Blough ER; Attenuation in

overload-induced skeletal muscle hypertrophy in obese insulin- resistant Zucker rat is

associated with hyperphosphorylation of AMP-activated protein kinase and dsRNA-

dependent protein kinase (PKR).

Nalabotu S, Takatsuki H, Kolli MB, Frost L, Yoshiyama S, Gadde M, Kakarla SK,

Kohama K, and Blough ER; Control of myosin enzyme activity by the reversible

alteration of protein structure.

ABSTRACTS

Presented several abstracts and posters at scientific meetings.

152

CLINICAL EXPERIENCE

Sep, 2010-Present Volunteer at Help for Animals Clinic, Huntington, WV

Oct, 2009-Mar, 2010 Volunteer at Proctorville Animal Clinic, Proctorville, OH

Oct, 2009- Jan, 2010 Volunteer at Equine Medical Center, Chesapeake, OH

July, 2003-Jun, 2005 Lead Veterinarian at Govt. Small animal clinic, Vizag, A.P,

India

July, 2002-Jun, 2003 Shelter Veterinarian, SPCA, Vizag, A.P, India

Jan, 2000-Jun, 2002 Veterinary Assistant Surgeon, Veterinary Polyclinic, A.P,

INDIA.

July, 1999-Dec, 1999 Intern, Veterinary Polyclinic, Hyderabad, A.P, INDIA.

Duties performed as a Veterinarian

Responsible for primary care including vaccination, prevention, diagnosis

and treatment of sick animals, and advising owners about nutrition and

management practices in an urban hospital setting which routinely dealt

with a mix of small (70%) and large (30%) animals.

Performed elective surgeries like spay, neuter, declawing, tail docking etc.

Performed reproductive soundness exams in dairy cattle, artificial

insemination, pregnancy diagnosis, handled dystocia cases and caesarian

section in needy situations.

Conducted farmer awareness camps about deworming, vaccination and

better management practices for livestock.

Trained technicians about animal handling, workplace safety, work

protocols, equipment handling and client interaction

Expertise:

Can perform dog spay in 15-20 minutes

Can perform dog neuter in 7-8 minutes

Can perform cat spay in 7-8 minutes

Can perform cat neuter in 5 minutes

Experienced in handling stainless steel suture material

Duties performed as volunteer in USA

Assisted veterinarians handle small animal medicine cases, learnt to

communicate with clients and prescribe appropriate medications.

153

Assisted with physical exams, obtaining and interpreting radiographs and

performing ultrasound exams.

Learned to analyze laboratory test results, interpret radiographic and

Ultrasound findings. Ability to operate both X-ray and Ultrasound

machines. Proficient in handling IDEXX and Vet Scan Laboratory

Systems.

Performed Urine and Fecal analysis.

Performed several spay and neuters, habituated to aseptic precautions,

anesthesia delivery (Induction to Recovery), vaccination and deworming

protocols.

Gained knowledge of record keeping and training of technicians.

HONORS AND AWARDS

Best veterinarian of the district (2002), Visakhapatnam, A.P, India

PROFESSIONAL MEMBERSHIPS

Member of AVMA

Member of Indian Veterinary Council

Eligibility to practice veterinary medicine in India.

Member of Andhra Pradesh Veterinary Association

License to practice veterinary medicine in the state of Andhra Pradesh, India.

RESEARCH SKILLS

Immunohistochemistry

Flourescent and Confocal Microscopy

Different staining procedures

Sample preparation from Laboratory animals

Protein Quantitation using Bradford assay

SDS gel electrophoresis

Immunoprecipitation and Westerns

Quantitation using Densitometry

DNA, RNA and miRNA isolation

DNA and Protein gel electrophoresis

PCR, RT-PCR

Cell culture

154

Small animal surgery

Handling of Laboratory animals

Cardiac Ischemia-Reperfusion Experiments in Rats and Mice

ELISA

COMPUTER SKILLS

Excellent experience with MS Office. (MS Word, Excel, PowerPoint,

Outlook and Access)

Excellent experience with statistical software – SigmaStat

Excellent experience with imaging software – Alpha Ease, Image J

Excellent experience with Adobe software (Acrobat, Photoshop), Endnote.

REFERENCES

Dr. Eric R. Blough, Ph.D.,

Assosciate Professor

Department of Biology

Director, Laboratory of Molecular physiology

Marshall University, Huntington, WV

E-mail: [email protected],

Phone: (304) 696-2708

(Master’s and PhD adviser)

Dr. Nalini Santanam, Ph.D.,

Professor and Cluster Coordinator

Dept of Physiology, Pharmacology and Toxicology

Joan C Edward School of Medicine

Marshall University, Huntington, WV

Phone: (304) 696-7321

E-mail: [email protected],

Dr. Todd L. Green, Ph.D.,

Associate Professor and

Director of Graduate studies, Biomedical Sciences Graduate programme

Dept of Pharmacology, Physiology and Toxicology

Phone: (304) 696-3531 Fax: (304) 696-7272

E-Mail: [email protected]