Understanding the Molecular Mechanisms of Post-Exercise ...

163
Understanding the Molecular Mechanisms of Post-Exercise Muscle Inflammation and Repair by Luke Vella BAppSc(Ex&SpSc)(Hons) Submitted in fulfilment of the requirements for the degree of Doctor of Philosophy Deakin University December 2016

Transcript of Understanding the Molecular Mechanisms of Post-Exercise ...

Understanding the Molecular Mechanisms of Post-Exercise Muscle

Inflammation and Repair

by

Luke Vella

BAppSc(Ex&SpSc)(Hons)

Submitted in fulfilment of the requirements for the degree of

Doctor of Philosophy

Deakin University

December 2016

lswan
Redacted stamp
lswan
Redacted stamp

Acknowledgements

I dedicate this thesis to my parents, Steve and Anne-Maree Vella, you are my support, my inspiration,

my family.

To my supervisory team, Aaron Russell, David Cameron-Smith and Rod Snow. Thank you for your

constant support, hard work, care and diligence. Your patience and understanding of my career

outside of my PhD has been enormously appreciated. Your passion and knowledge in your

respective fields is something that I will always admire and aspire to.

To my co-authors, James Markworth, Jonathan Peake, Rao Maddipati, Gøran Paulsen, Truls Raastad,

Marissa Caldow, Petra Graves, Amy Larsen, Paul Della Gatta and Daniella Tassoni. You have all

contributed significantly to my research and to this thesis. Thank you for your time, dedication and

support over the past 8 years.

To Dr. Andrew Garnham. It was a pleasure working with you through the exercise trials. Your

expertise is unrivalled and I hope our paths will cross again in the future.

To Dr. Sean Williams and Professor John Reynolds. Thank you for all of your support with my

statistical analysis, I cannot express how lost I would have been without your guidance. Your

patience and willingness to help me through the process was a saving grace.

To Laura. My best friend, my never ending support, my inspiration and soon-to-be, my wife. I could

write another thesis on how much I treasure your love and support. Thank you for being all of those

things and so much more. You have kept me sane and grounded throughout this entire process.

Finally to my family. My brother Mark, my Mum Anne-Maree and my Dad Steve. You have provided

me with every opportunity that a person could possibly wish for. You are the first people I want to

talk to when things go well, and the first people I turn too when things go wrong. Your love,

generosity, selflessness and modesty is something that I aspire to every day of my life.

List of publications

Vella L, Markworth JF, Paulen G, Raastad T, Peake JM, Snow RJ, Cameron-Smith D, Russell

AP: Ibuprofen ingestion does not affect markers of post-exercise muscle inflammation.

Frontiers in Physiology, March 2016, 7:86.

Vella L, Markworth JF, Peake JM, Snow RJ, Cameron-Smith D, Russell AP: Ibuprofen

supplementation and its effects on NF-κB activation in skeletal muscle following resistance

exercise. Physiological reports, September 2014, 2:10.

Vella L, Caldow MK, Larsen AE, Tassoni D, Della Gatta PA, Gran P, Russell AP, Cameron-Smith

D: Resistance exercise increases NF-κB activity in human skeletal muscle. American Journal

of Physiology. Regulatory, Integrative and Comparative Physiology, March 2012, 302:6.

Table of Contents

LIST OF FIGURES .......................................................................................................................... I

LIST OF TABLES .......................................................................................................................... III

ABBREVIATIONS ........................................................................................................................IV

CHAPTER 1 – LITERATURE REVIEW ............................................................................................ 1

1.1 Introduction: ....................................................................................................................... 2

1.2 Exercise-induced inflammation: ......................................................................................... 3

1.3 Anti-inflammatory treatments and muscle recovery: ........................................................ 6

1.3.1. Prostaglandin response to exercise. ............................................................................ 7

1.3.2. The effect of NSAIDs on muscle inflammation and recovery. ..................................... 7

1.4 Resolution of exercise-induced inflammation: .................................................................. 9

1.5 Transcriptional regulation of exercise-induced inflammation: ........................................ 12

1.5.1 Nuclear Factor-kappa B: ........................................................................................ 13

1.5.2 NF-κB and inflammation: ................................................................................... 15

1.5.3 NF- κB and skeletal muscle myogenesis: ........................................................... 15

1.5.4 NF- κB and exercise: ........................................................................................... 17

1.6 Summary: .......................................................................................................................... 18

1.7 General hypothesis: .......................................................................................................... 19

1.8 Specific Aims: .................................................................................................................... 21

1.9 Hypothesis: ....................................................................................................................... 21

CHAPTER 2: .............................................................................................................................. 22

2.1 Abstract: ............................................................................................................................. 23

2.2 Introduction: ...................................................................................................................... 24

2.3 Methods: ............................................................................................................................ 27

2.3.1 Participants: ................................................................................................................ 27

2.3.2 Ethics approval: ........................................................................................................... 27

2.3.3 Familiarization: ............................................................................................................ 28

2.3.4 Experimental Procedures: ........................................................................................... 28

2.3.5 Standardized meals: .................................................................................................... 28

2.3.6 NSAID Administration: ................................................................................................ 29

2.3.7 Sample collection: ....................................................................................................... 29

2.3.8 Immunohistochemistry: .............................................................................................. 30

2.3.9 Biochemical assays: ..................................................................................................... 31

2.3.10 Muscle soreness assessment: ................................................................................... 31

2.3.11 Statistics: ................................................................................................................... 32

2.4 Results: ............................................................................................................................... 33

2.4.1 Muscle Soreness: ......................................................................................................... 33

2.4.2 Immunohistochemistry: .............................................................................................. 33

2.4.3 Serum proteins: ........................................................................................................... 35

2.4.4 Correlation analysis: .................................................................................................... 36

2.5 Discussion: ......................................................................................................................... 38

2.6 Conclusion: ......................................................................................................................... 40

2.7 Acknowledgements:........................................................................................................... 41

2.8 Disclosures: ........................................................................................................................ 41

CHAPTER 3: .............................................................................................................................. 42

3.1 Abstract: ............................................................................................................................. 43

3.2 Introduction: ...................................................................................................................... 44

3.3 Methods: ............................................................................................................................ 47

3.3.1 Subjects: ...................................................................................................................... 47

3.3.2 Ethics approval: ........................................................................................................... 47

3.3.3 Familiarization: ............................................................................................................ 47

3.3.4 Experimental design: ................................................................................................... 48

3.3.5 Muscle biopsy procedure: ........................................................................................... 48

3.3.6 Liquid chromatography-mass spectrometry: .............................................................. 48

3.3.7 Statistics: ..................................................................................................................... 49

3.4 Results: ............................................................................................................................... 50

3.4.1 Lipidomic analysis:....................................................................................................... 50

3.4.1.1 Cyclooxygenase pathway: .................................................................................... 50

3.4.1.2 Lipoxygenase pathway: ........................................................................................ 52

3.4.1.3 Epoxygenase pathway: ......................................................................................... 55

3.5 Discussion: ......................................................................................................................... 57

3.6 Conclusion: ......................................................................................................................... 61

3.7 Acknowledgments: ............................................................................................................ 61

3.8 Disclosures: ........................................................................................................................ 61

CHAPTER 4: .............................................................................................................................. 63

4.1 Abstract: ............................................................................................................................. 64

4.2 Introduction: ...................................................................................................................... 65

4.3 Methods: ............................................................................................................................ 68

4.3.1 Participants: ................................................................................................................ 68

4.3.2 Ethics approval: ........................................................................................................... 68

4.3.3 Familiarization and strength testing: .......................................................................... 69

4.3.4 Experimental procedures: ........................................................................................... 69

4.3.5 NSAID administration: ................................................................................................. 70

4.3.6 Sample collection: ....................................................................................................... 70

4.3.7 Protein extraction and quantification: ........................................................................ 71

4.3.8 RNA extraction and RT-PCR: ........................................................................................ 71

4.3.9 Nuclear extraction and Transcription Factor (TF) Assay: ............................................ 72

4.3.10 Statistics: ................................................................................................................... 73

4.4 Results: ............................................................................................................................... 74

4.4.1 Protein expression: ..................................................................................................... 74

4.4.2 Electrophoretic mobility shift assay: ........................................................................... 76

4.4.3 RT-PCR: ........................................................................................................................ 77

4.5 Discussion: ......................................................................................................................... 79

4.6 Conclusion: ......................................................................................................................... 81

CHAPTER 5 – GENERAL DISCUSSION AND FUTURE DIRECTIONS: ............................................ 83

5.1 – Introduction: ................................................................................................................... 83

5.2 – Summary of results: ........................................................................................................ 84

5.3 – Future directions: ............................................................................................................ 85

5.4 – Concluding remarks: ....................................................................................................... 87

REFERENCE LIST: ...................................................................................................................... 89

Appendix A: ............................................................................................................................ 122

Appendix B: ............................................................................................................................ 129

Appendix C: ............................................................................................................................ 145

I

LIST OF FIGURES

Figure 1.1: A schematic of the process of acute exercise-induced muscle damage,

inflammation and repair.

Figure 1.2: NF-κB - a central regulator of stress signaling pathways.

Figure 1.3: Classical and alternative NF-κB signaling pathways.

Figure 2.1: Subjective rating for delayed onset muscle soreness (DOMS). This figure has

been adapted from Vella et al. (2014).

Figure 2.2: Immunohistochemistry data for muscle cells staining positive for WBC cell

surface markers.

Figure 2.3: Immunohistochemical analysis of skeletal muscle samples following a bout of

resistance exercise.

Figure 2.4: Blood derived proteins creatine kinase and myoglobin acute resistance exercise

Figure 3.1: Metabolites of the cyclooxygenase pathway derived from arachidonic acid.

Figure 3.2: Metabolites of the lipoxygenase pathway derived from arachidonic acid.

II

Figure 3.3: Metabolites of the lipoxygenase pathway derived from docosahexaenoic acid.

Figure 3.4: Metabolites of the epoxygenase pathway derived from arachidonic acid.

Figure 3.5: Metabolites of the epoxygenase pathway derived from linoleic acid.

Figure 4.1: Protein expression of NF-κB subunits p65, p50, p52 and cREL.

Figure 4.2: NF-κB subunits p65 (A) and p50 (B) binding to nuclear protein.

Figure 4.3: RT-PCR analysis of NF-κB target genes IL-6 (A), IL-8 (B), MCP-1 (C), TNF-α (D) and

COX-2 (E) in skeletal muscle cDNA.

III

LIST OF TABLES

TABLE 2.1 SUBJECT CHARACTERISTICS AND STRENGTH TESTING DATA.

TABLE 2.2 SPEARMAN RANK CORRELATION ANALYSIS.

TABLE 4.1 SUBJECT CHARACTERISTICS AND STRENGTH TESTING DATA.

TABLE 4.2 RT-PCR PRIMER SEQUENCES

IV

ABBREVIATIONS

1RM 1 repetition maximum

AA Arachidonic acid

ATP Adenosine triphosphate

BMI Body mass index

BSA Bovine serum albumin

CK Creatine kinase

COX Cyclooxygenase

CYP Cytochrome P450

DHA Docosahexaenoic acid

DiHETrE Vicinal diol dihydroxyeicosatrienoic acid

DOMS Delayed onset muscle soreness

EMSA Electrophoretic mobility shift assay

EPA Eicosapentaenoic acid

EpETrE Epoxyeicosatrienoic acid

HDoHE Monohydroxylated- docosahexaenoate

HETE Hydroxyeicosatetraenoic acids

IBU Ibuprofen

IGF-BP1 Insulin-like growth factor binding protein 1

IGF-BP2 Insulin-like growth factor binding protein 2

IGF-II Insulin-like growth factor II

IKK Inhibitor of κB kinase

IL-6 Interleukin-6

IL-8 Interleukin-8

iMVC Isokinetic maximal voluntary contraction

IκB Inhibitor of κB

LC-MS Liquid chromatography-mass spectrometry

LOX Lipoxygenase

LSD Least significant difference

LT Leukotriene

LX Lipoxin

MAPK Mean arterial pressure kinase

MCP-1 Monocyte chemoattractant protein-1

MHC Myosin heavy chain

MPO Myeloperoxidase

MYO Myoglobin

n-3 Omega-3

n-6 Omega-6

V

NF-κB Nuclear factor-kappa B

NSAID Non-steroidal anti-inflammatory drug

PCR Polymerase chain reaction

PD Protectin

PDGF Plasma-derived growth factor

PG Prostaglandin

PLA Placebo

PMN Polymorphonuclear neutrophils

PUFA Polyunsaturated fatty acid

Rv Resolvin

SEM Standard error of the mean

SPM Specialized pro-resolving mediator

TGF-β Transforming growth factor-β

TMB Tetramethylhylbenzidine

TNF-ɑ Tumour necrosis factor-ɑ

TX Thromboxane

VEGF Vascular endothelial growth factor

YY1 Ying Yang1

1

CHAPTER 1 – LITERATURE REVIEW

2

1.1 Introduction:

Skeletal muscle is a remarkably heterogeneous tissue with a vast capacity to adapt and

respond to its external environment [1-4]. It is therefore not surprising that the intense

muscular contractions common to resistance exercise results in a marked increase in muscle

size, commonly termed hypertrophy [5, 6]. However, following most bouts of intense exercise,

there is often considerable muscle damage, inflammation and muscle soreness. Human studies

exploring the physiological mechanisms of exercise-induced muscle damage date back to the

beginning of the 20th century [7]. Early works with muscle preparations and animal models

demonstrated that physical exercise causes damage to muscle cells including the mechanical

disruption of sarcomeres and adjacent cellular membranes, and disturbed excitation

contraction coupling and calcium signalling. [8-15]. Pioneering work using human skeletal

muscle biopsy samples reported muscle damage at the ultrastructural level and changes in

circulating leucocytes [16-22]. These studies identified a role for inflammatory white blood

cells in post-exercise muscle recovery. Modern molecular biology has superimposed additional

layers of complexity by, identifying functional roles for protein and lipid derivatives including

cytokines, acute phase proteins and eicosanoids, in exercise-induced inflammation. Whilst

precise cellular and molecular signalling pathways remain poorly understood, post-exercise

muscle recovery occurs according to a well-described temporal scheme: (1) tissue injury via

mechanical and metabolic stress, (2) release of vasoactive and chemotactic substances from

damaged tissue, (3) migration and adhesion of systemic leucocytes to the injury site, and (4)

tissue repair and adaptation (Figure 1). Research efforts over the last few decades have sought

to understand the physiological significance of acute post-exercise inflammation and its

functional role in muscle recovery and adaptation. Developing an understanding of the

molecular mechanisms that regulate this phenomenon may permit the development of

therapeutic treatment strategies to manipulate selective events of muscle inflammation and

optimise the cellular processes of muscle repair.

3

Figure 1.1: A schematic of the process of acute exercise-induced muscle damage, inflammation and

repair. This figure was adapted from Pillon et al. and demonstrates the interaction between skeletal

muscle and both local and systemic immune cells [23].

1.2 Exercise-induced inflammation:

It appears that cellular events occurring early in exercise-induced muscle damage engage a

biologically active and tightly regulated acute inflammatory process that is of critical

importance to post-exercise muscle recovery. While the complexity of inflammation is

currently acknowledged herein, the fundamental objective of any inflammatory program is to

repair injury and restore tissue function. Historically, the paradigm that exercise-induced

muscle damage is linked to an acute inflammatory response has been driven by reports from

various animal reports and in-vitro study designs, and has been extensively reviewed [9, 12, 13,

24-30]. Because of the various physiological and anatomical differences between species, and

the extent of muscle damage that can be caused in in-vitro research models, findings from

these studies may or may not be applicable to voluntary physical activity in humans.

Localized inflammation is initiated by damage to the extracellular matrix, increasing the

permeability of the sarcolemmal membrane, causing a rapid extravasation of fluid and blood-

borne polymorphonuclear neutrophils (PMNs) into the damaged tissue [31-33]. It is well

accepted that neutrophils play a critical role in acute inflammation, removing necrotic tissue

and cellular debris via a process termed phagocytosis [31, 34]. Phagocytosis triggers a

4

respiratory burst and subsequent release of cytotoxic granulocyte products, reactive oxygen

species and inflammatory cytokines, which cause further tissue damage and exacerbate the

inflammatory response [35-38]. Neutrophil infiltration appears to peak between 3-24 h post-

exercise. Interestingly this time point is associated with a rise in histological and functional

markers of muscle damage. This suggests that neutrophil function may lead to secondary

tissue injury, a phenomenon termed delayed onset muscle soreness (DOMS) [8, 39-43]. Peak

accumulation of PMNs is overlapped in time by the accumulation of monocyte/macrophages.

These are highly versatile cell types whose function depends largely on the extracellular

environment [44, 45]. Two sub-types of macrophages exist within skeletal muscle, and perform

multiple and largely disparate functions within the contraction-induced inflammatory cascade

[25, 34, 46-48]. Classically activated, or M1 macrophages, appear to peak between 12 and 24 h

post exercise [49] and are most prominent within necrotic muscle fibres [50, 51]. The

distribution of M1 cells is consistent with their capability of phagocytosis [52]. This precedes a

shift in macrophage phenotype to an alternatively activated, or M2 macrophage. This shift in

phenotype coincides with a change from the proliferative stage to the early differentiation

stage of myogenesis, and substantiates a long suspected concept that M2 macrophages

regulate key pathways in muscle regeneration [25, 33, 47, 53-60]. These findings rationalize

the concept that cellular events occurring early in the acute inflammatory cascade, trigger a

series of cellular processes that are a key regulatory component of muscle regeneration and

tissue repair.

More recent human research identified two key concepts that have challenged the paradigm

of exercise-induced muscle damage, DOMS, and muscle regeneration. Firstly, a tissue may be

influenced by pro-inflammatory signaling molecules, including cytokines and eicosanoids, in

the absence of inflammatory cell invasion. Subsequently, humans can experience symptoms of

exercise-induced muscle damage without presenting with cellular signs of inflammation [61-

65]. Radionuclide imaging techniques have confirmed large changes in circulating leucocytes

(primarily neutrophils) immediately following exercise [63, 66]. However, these techniques

give no information about the infiltration of leucocytes into skeletal muscle tissue.

Investigations of exercise-induced muscle inflammation that explore the infiltration of

leucocytes in to skeletal muscle tissue are relatively scarce, and conflicting results have often

5

been attributed to variations in exercise protocols, muscle sampling procedures, individual

responsiveness to exercise stress. Additionally, the use of non-specific nomenclature to

identify and describe muscle damage and inflammation [17, 19, 67-74] has also created some

confusion in the field. The main criticism of these studies is the risk of the muscle biopsy

procedure triggering local inflammatory responses that could confound the effects of the

exercise protocol. Muscle biopsy procedures have been shown to cause an increase in the

activity of mean arterial pressure kinase (MAPK) [75] and the expression of leucocyte specific

antigens [62], while impairing adenosine triphosphate (ATP) and glycogen re-synthesis [76].

Alternatively, indirect proxy measures of muscle damage and inflammation including

subjective DOMS, swelling and serum measures of blood-borne proteins have been used,

however these markers do not accurately reflect the extent of cellular muscle damage and

show poor correlation with each other [20, 77-82]. In order to determine a functional role for

acute inflammation in exercise-induced muscle damage, further research including a non-

exercise control group, and measuring direct markers of cellular inflammation are required.

The presence of a post-exercise inflammatory response remains well accepted, however its

relationship with myofibrillar disruption, cellular damage and muscle soreness remains

speculative [19, 65, 83, 84]. A review from Paulsen et al. [85] suggested that when intracellular

damage exceeds a specific threshold, leucocytes infiltrate the damaged tissue and this

manifests itself as a reduction in muscle force generating capacity. A reduction in muscle force

generating capacity of greater than 20% appears necessary to initiate leucocyte accumulation

in skeletal muscle tissue, however results remain inconsistent [19, 84, 86-92]. Studies reporting

a reduction of muscle force generating capacity greater than 50%, consistently resulted in an

increase in intramuscular leucocyte infiltration [69, 70, 84, 93-96].

Immunohistochemical staining of leucocyte specific antigens has become the gold standard for

the temporal profiling of the appearance of leucocyte populations in human muscle following

exercise. While animal and in-vitro studies present a highly predictable sequence of cellular

events in acute inflammation, these findings have yet to be replicated in human in-vivo

exercise clinical trials. Pioneering works from Paulsen et al. [84] attempted to profile the time

6

course appearance of leucocyte populations by staining muscle cells for CD16 (primarily a

neutrophil marker) and CD68 (primarily a monocyte/macrophage marker). This study showed

increased numbers of both CD16 and CD68 localised to the endomysium and perimysium as

soon as 0.5 h post-exercise and persisting until 4 d post-exercise [84]. Further, this study

established a strong correlation between the individual changes in muscle function and the

accumulation of radiolabelled leucocytes, providing further justification for a role of exercise-

induced inflammation in delaying the restoration of muscle function post-exercise. The

appearance of monocyte/macrophage populations has been a consistent histological finding

and appears common in both early stage [86, 88, 89, 92, 93, 97] and late stage inflammation

[32, 70, 98]. Among those publications exclusively examining neutrophil infiltration only two

studies have identified an increase in both early stage [68, 84] and late stage inflammation [84,

98]. Further research profiling the time-course of leucocyte recruitment and infiltration in

human subjects is required to understand the complexity of the exercise-induced inflammation

response.

1.3 Anti-inflammatory treatments and muscle recovery:

Non-steroidal anti-inflammatory drugs (NSAIDs) are a commonly used as a treatment strategy

in exercise and sports medicine to assist with recovery from exercise-induced inflammation,

particularly following soft-tissue injury [99]. NSAIDs inhibit the cyclooxygenase (COX-1 and -2)

enzymes and consequently the formation prostanoids derived from mobilised free intracellular

arachidonic acid (AA) [100]. The COX pathway converts AA to prostaglandin G2 (PGG2), and

subsequently catalyses the reduction of PGG2 to form PGH2 [101]. Various synthase enzymes

convert PGH2 to form five primary prostanoids that play a diverse role in acute inflammation,

including; thromboxane A2, PGD2, PGF2ɑ, PGE2 and PGI2. These prostaglandin species are often

ascribed to the cardinal signs of acute inflammation through their effects on local blood flow,

vascular permeability, leucocyte infiltration, and triggering sensations of pain [102, 103].

However, more recent information from both animal [104-106] and human research [87, 107,

108], suggests a role for prostaglandin species in skeletal muscle regeneration.

7

1.3.1. Prostaglandin response to exercise.

Research in to the prostaglandin response to muscle loading and exercise-induced muscle

damage has produced equivocal findings that appear to be influenced by the nature of the

exercise stimulus. Early studies implementing a prolonged submaximal exercise protocol

demonstrated heightened levels of various circulating prostaglandin species including PGE2

[109-112], PGF2ɑ [102, 109], PGI2 [113-116] and thromboxane [113, 116]. Fewer studies have

investigated the prostaglandin response to muscle damaging resistance exercise protocols. The

limited studies available demonstrated an increase in circulating PGE2 and 13,-14-dihydro-15-

keto-PGF2ɑ following both stretch-shortening cycling exercise [117] and traditional resistance

exercise [118-120]. No such effect was observed in isolated eccentric knee extension [121, 122]

and elbow extension exercise [123-125], or downhill running protocols [126]. Similarly, few

studies have attempted to profile intramuscular prostaglandin responses to exercise-induced

muscle damage. Skeletal muscle cells express both COX-1 and -2. Few studies have reported an

increase in skeletal muscle COX mRNA expression and enzymatic activity during exercise

recovery [127-130], while others have found no change [87, 131]. Eccentric resistance exercise

triggers an increase in intramuscular PGF2ɑ at 24 h post-exercise [132], while a more traditional

bout of resistance exercise increases PGF2ɑ in muscle microdialysates at 5-6 and 8-9 h post-

exercise, with no increase in PGE2 [133]. Likewise, an incremental cycling based intervention

triggered an increase in PGE2 concentration during exercise, measured using microdialysis,

however an intermittent plantar flexion protocol elicited no such response [134, 135]. Further,

unilateral arm exercise including 70 maximal eccentric contractions, achieved no change in

PGE2 levels in tissue dialysate [93]. The reason for such conflicting findings remains unclear,

however may relate to differences in the exercise protocol, specifically in the intensity of

muscle load, or the volume of active muscle mass in the respective exercise trials.

1.3.2. The effect of NSAIDs on muscle inflammation and recovery.

Administration of non-selective NSAIDs (e.g. ibuprofen) effectively blocks the exercise-induced

rise in prostaglandins in both skeletal muscle [132, 136, 137] and peripheral blood [102].

Therefore, NSAID treatment has been used as a research model to explore the physiological

role of prostaglandin species in exercise-induced inflammation, and the biological link between

acute inflammation and skeletal muscle regeneration. Research into the effects of NSAIDs on

8

exercise-induced muscle damage and inflammation has produced largely controversial

findings. Many studies have explored the short term effects of NSAID treatment on indirect

markers of muscle damage and inflammation and produced disparate findings for an effect on

muscle swelling [93, 138, 139], circulating creatine kinase [42, 119-121, 140-146], restoration

of muscle function [42, 93, 122, 138, 140, 142, 144, 146-150] and subjective muscle soreness

[42, 71, 87, 93, 107, 119, 138-158]. While these markers often represent the symptoms of

exercise-induced muscle damage, many of these parameters have a relatively weak association

with structural muscle damage or, more specifically, cellular inflammation. In animal models of

acute muscle damage, treatment with NSAIDs blunts the infiltration of leucocytes into muscle

tissue [104, 159], however this finding has not been replicated in human subjects [147, 160].

Oral consumption of both ibuprofen, and acetaminophen, an analgesic also known as

paracetamol, had no effect on skeletal muscle macrophage infiltration at 24 h following an

eccentric exercise protocol [160]. Similarly treatment with naproxen, another non-selective

NSAID, had no effect on the infiltration of leucocyte common antigen positive cells following a

unilateral, isotonic resistance exercise protocol [147]. Interestingly, Paulsen et al., (2010)

suggested a blunting effect of the COX-2 specific celecoxib following maximum eccentric

muscle contractions [93]. This research identified a tendency for higher monocyte/macrophage

numbers in subjects within the placebo group, and within those subjects who were identified

as ‘high-responders’ to the exercise protocol based on the number of inflammatory leucocytes

[93]. Although this is not a particularly robust finding, it has led to the hypothesis that NSAIDs

may influence leucocyte infiltration in skeletal muscle when a sufficiently strong and early

inflammatory reaction is present [93]. This hypothesis would suggest that the intensity of the

exercise stimulus and the consequent muscle-damage response would be largely influential in

determining the effect of a pharmacologically based anti-inflammatory intervention.

Experimental evidence also indicates that NSAID administration can influence the regenerative

capacity of skeletal muscle following exercise-induced muscle damage. The non-selective COX

inhibitor ibuprofen, blunted skeletal muscle protein synthesis [107], while local intramuscular

infusion of indomethacin inhibited satellite cell proliferation following maximum eccentric

exercise protocols [87]. Similarly, oral administration of indomethacin attenuated the exercise-

induced increase in satellite cell number following an endurance running protocol. Further,

9

treatment with ibuprofen inhibited early translational signaling responses involved in post-

exercise muscle hypertrophy following a bout of traditional resistance exercise [161]. These

findings support earlier in-vitro research models that implicate a role for prostaglandin species

in stimulating skeletal muscle adaptive pathways [162, 163]. However, follow up studies using

selective COX-2 specific NSAID celecoxib showed no effect on the exercise-induced increase in

skeletal muscle protein turnover [130] or satellite cell number [93] following an eccentric

exercise protocol. These findings present two potential alternatives. Firstly, despite the well-

established role for the inducible COX-2 enzyme in the animal muscle reparative response to

acute damage [104-106, 111, 164], the constitutively active COX-1 may play an important

function in the human adaptive response to exercise. Alternatively, NSAID administration may

have the capacity to influence alternative signalling pathways within the exercise-induced

inflammation-tissue repair paradigm. Markworth et al. employed a targeted lipidomics

approach to characterize temporal changes in the human peripheral blood fatty acyl lipid

mediator profile in response to acute resistance exercise [102]. This research demonstrated

that ibuprofen administration has the capacity to influence the appearance of a series of lipid-

derived mediators that have the potential to engage a biologically active inflammatory

resolution programme that may provide a crucial physiological link between inflammatory and

adaptive signaling pathways.

1.4 Resolution of exercise-induced inflammation:

Historically, the label of inflammation has been ascribed to a distinct group of symptoms

including heat, redness, swelling, pain and a loss of function [165]. The resolution of

inflammation was previously thought to be a passive process whereby chemotactic gradients

were thought to dissipate and the exodus of inflammatory leucocytes through lymphatic

drainage enabled muscle tissue to return to homeostatic function. However, the discovery of a

novel class of lipid-derived mediators has established the resolution of inflammation as an

active biochemical and metabolic process. The profiling of these bioactive lipids was pioneered

by Serhan et al. [166-174] and represents an area of critical importance in understanding the

biological significance of the inherent self-resolving nature of acute skeletal muscle

inflammation.

10

Lipid mediators are biosynthesized endogenously from numerous polyunsaturated fatty acid

(PUFA) precursors including essential omega-3 fatty acids (n-3) eicosapentaenoic (EPA) and

docosahexaenoic acid (DHA), and major omega-6 (n-6) arachidonic acid (AA). Under basal

conditions PUFA substrates remain esterfied within cell membrane phospholipids. In response

to injury or inflammatory insult hundreds of distinct lipid species can be synthesized via 3

major enzymatic pathways: (1) cyclooxygenase, (2) lipoxygenase, and (3) epoxygenase which is

catalysed by cytochrome P450 (CYP) [101]. The development of mass spectrometry (MS)-based

lipidomic profiling has identified and quantified large numbers of pro-inflammatory, pro-

resolution and anti-inflammatory mediators, and enabled the sequential profiling of lipid

species in a variety of acute inflammatory models. The majority of research has focussed on

classic prostaglandin and leuokotriene species which are pro-inflammatory in nature and

derived from omega-6 AA. These lipids play a diverse role in stimulating acute inflammation by

controlling local blood flow, cytokine production, cell membrane permeability and leucocyte

infiltration [102]. More recently, a secondary class of eicosanoids also generated from AA,

termed lipoxins [172, 175, 176], as well as newly identified EPA (E-Series) and DHA (D-Series)

derived resolvins, protectins [166, 169-171, 177, 178], play dual anti-inflammatory and pro-

resolution functions in acute inflammation. These endogenous mediators possess potent

immunoregulatory actions via several pathways including the inhibition of pro-inflammatory

cytokines and the chemotaxis neutrophils, and stimulating the recruitment of monocytes and

the nonphlogistic phagocytosis of apoptotic neutrophils [175, 177-182]. Evidence from in-vitro

models suggests that the phagocytosis of apoptotic neutrophils can trigger a phenotypic shift

in macrophages from the classical M1 macrophage, to the alternative M2 macrophage [181].

This process may suppress the release of pro-inflammatory agents and increase the

macrophage release of anti-inflammatory mediators such as transforming growth factor-β

(TGF-β) and vascular endothelial growth factor (VEGF) [44, 183].

Lipoxins, resolvins and protectins are formed during inflammatory transcellular interactions,

involving the sequential actions of two or more LOX and/or COX enzymes. During inflammation

cell-cell interactions between platelets, leucocytes and resident tissue cells facilitates the

transcellular biosynthesis of unique lipid derivatives [101]. For example, LX biosynthesis

involves cellular interactions between 5-LOX expressing PMNs with 12-LOX expressing platelets

11

or 15-LOX expressing M2 monocytes [184]. The complex signalling network that exists within

damaged skeletal musculature remains poorly understood, and it appears from other models

of acute inflammation that the temporal regulation of lipids and their role in inflammation is

likely to be cell-type and stimulus specific.

Our understanding of pro-resolving lipid mediator circuits has been limited to in-vitro assays

utilizing blood borne immune cell populations and pharmacologically induced models of acute

inflammation. The first study to explore the time-course appearance of lipid mediators

implemented a TNF-ɑ-stimulated model of acute inflammation in the murine air pouch [185].

Within this model, early formation of LTB4 and PGE2 at the onset of inflammation was

succeeded by a class-switching of eicosanoids to LXA4 which was concurrent with spontaneous

inflammatory resolution. During this process, interactions between inflammatory and host

tissue cells enabled the biosynthesis of resolution mediators [185]. Human PMN exposed to

PGE2 switched eicosanoid biosynthesis from predominately LTB4 to a LXA4 product, establishing

a pathway termed lipid mediator class switching. Alternatively, in a model of zymosan-A

stimulated murine peritonitis, the onset of inflammation was characterized by a concomitant

increase in LTB4 and LXA4 followed by a late appearance of PGE2 at the onset of resolution

[186]. The discrepancies in the temporal patterns of eicosanoid generation likely reflect

differences in the nature of the experimental models (microbial-initiated vs wound model)

and/or the stimulus specific signalling pathways (zymosan A vs TNF-ɑ).

In the first study to explore the appearance of lipid derived resolution mediators in an in-vivo

exercise model, maximal physical exertion on a cycle ergometer caused a rapid increase in the

urinary excretion of LXA4 immediately post-exercise [187]. These findings led the author to

suggest that the enhanced LX biosynthesis may represent a protective pro-resolving

mechanism programmed in to the early stages of acute inflammation to act as a self-regulatory

defence against exercise-induced cellular stress [187]. More recently Markworth et al. [102]

undertook an unbiased metabololipidomic profiling of human peripheral blood serum samples

obtained during the first 24 h of muscle recovery following an acute bout of unaccustomed

resistance exercise. This study showed an increase in key prostaglandin, leukotriene, lipoxin

12

and E-series resolvin species during the early stages of acute post-exercise inflammation (0-3

h). Subsequently D-series resolvins and protectins, along with 15-LOX derivatives and some

prostaglandin metabolites (6-keto-PGF1ɑ and 13,14dh-15kPGE2) remained elevated within the

circulation following 24 h of recovery [102]. Collectively these results indicate that pro-

resolving lipid mediator biosynthesis is increased in humans in response to an exercise stress

and the concept of an active inflammatory resolution model must be considered. Distinct

classes of pro-resolution lipid mediators appear to exhibit specific temporal patterns

throughout the time-course of post-exercise muscle recovery. Therefore, future research

needs to establish the profile of resolution mediators, and the transcriptional mechanisms that

regulate this pathway, in skeletal muscle tissue in models of exercise-induced inflammation.

1.5 Transcriptional regulation of exercise-induced inflammation:

To understand the link between inflammatory and regenerative processes requires the

identification of molecular signalling pathways coupling cellular stress with tissue adaptation.

NF-κB is a pleiotropic transcription factor that acts as a central regulator of stress signaling

pathways by mediating a wide array of cellular responses including immune cell recruitment,

cellular survival, proliferation and differentiation (Figure 2) [188-195]. The regulation of NF-κB

is implicated in many of the physiological processes that occur in response to tissue injury,

specifically: (1) the onset of inflammation, (2) the resolution of inflammation and (3) skeletal

muscle myogenesis.

13

Figure 1.2: NF-κB acts as a central regulator of stress signaling pathways and is involved in a diverse

number of stress signaling pathways. The function of NF-κB within cellular physiology depends on the

inciting stimulus and the tissue type.

1.5.1 Nuclear Factor-kappa B:

In mammalian tissue the Rel/NF-κB family of eukaryotic transcription factors is composed of

five structurally related subunits, RelA (p65), cREL, RelB, NF-κB1 (p50) and NF-κB2 (p52) [191,

192]. These proteins bind to form hetero- and homodimers that produce diverse combinations

of dimeric complexes [196, 197]. Each dimer possesses different DNA binding specificities, thus

different complexes can induce distinct signaling pathways specific to the activating stimulus

and cell type [196, 197]. Under basal conditions NF-κB remains sequestered within the

cytoplasm in an inactive complex bound to the inhibitor protein IκB [190, 193, 198]. There are

seven mammalian IκBs; IκBα, IκBβ, IκBε and IκBγ, BCL-3 and precursor proteins p100 and p105

that interact with specific NF-κB dimers. Upon stimulation the IκB kinase (IKK) complex

regulates the degradation of IκB proteins via phosphorylation and subsequent

polyubiquitination. The IKK complex is composed of two catalytic subunits IKK-α and IKKβ and a

regulatory subunit IKK-γ [192]. The activation and subsequent nuclear translocation of NF-κB

dimeric complexes occurs via two well described mechanisms; the classic and alternative NF-κB

14

signaling pathways. Briefly, activation signals are channelled through intracellular adaptive

proteins which permit receptor induced activation of IKK. Within the classical activation

pathway, the activation of IKKβ and IKK-γ induces the phosphorylation and consequent

degradation of IκBα and IκBβ via the proteosome ubiquitination pathway [192, 199].

Dissociation from IκB permits the nuclear localization of NF-κB subunits p50 and p65 to

transcriptionally regulate the activation of specific target genes (Figure 3). Alternative NF-κB

signaling proceeds through the activation of IKK-α which triggers the phosphorylation and

partial proteolysis of precursor protein p100 to form the p52 subunit via a co-translation

mechanism [194]. Once processed, p52 forms a dimeric complex with RelB which, upon

activation, translocates to the nucleus to regulate the transcription of genes that may be

distinctly different from those targeted by the classical pathway [194] (Figure 3).

Figure 1.3: Classical and alternative NF-κB signaling pathways [194].

15

1.5.2 NF-κB and inflammation:

NF-κB regulates the expression of various genes that play a pivotal role in both acute and

chronic inflammatory conditions. In response to a diverse range of pro-inflammatory stimuli

including; cytokines, oxidative and thermal stress, ischemia and apoptotic mediators, NF-κB

induces pro-inflammatory genes including cytokines [200], chemokines [192], vascular and

intracellular adhesion molecules [201, 202], and acute phase proteins [203]. NF-κB also plays a

role in the promoting the cessation of an inflammatory program via the inhibition of neutrophil

apoptosis [204]. Dysregulation of apoptosis inevitably leads to the disintegration of

extravasated neutrophils. This in turn exposes surrounding tissue to large quantities of

histotoxic products causing further damage and prolonging the inflammatory response [183].

Not surprisingly chronic up regulation of NF-κB is implicated in the pathogenesis of several

inflammatory myopathies [205, 206] establishing NF-κB as a prospective target for

manipulation in the treatment of chronic inflammatory disorders [207]. However recent

research has identified a role for NF-κB in the active resolution program of acute inflammation

[208-210]. Homodimers of the alternative NF-κB activation pathway (p50 subunit) which lack a

transactivation domain, are associated with the expression of anti-inflammatory genes, the

inhibition of pro-inflammatory stimuli and the induction of apoptosis [210-212]. In a model of

rodent carageenin-induced pleurisy NF-κB activation was biphasic in nature [210]. Early

activation of the classical NF-κB pathway coincided with the onset of inflammation and the

expression of pro-inflammatory cytokines, while a secondary activation of the alternative NF-

κB pathway comprising p50 homodimers corresponded with the resolution of inflammation

[210].

1.5.3 NF- κB and skeletal muscle myogenesis:

The role of NF-κB in this process remains enigmatic and appears to be specific to the duration

of NF-κB activation, the inducing stimuli and the dimeric complex activated. Accumulating

evidence suggests that NF-κB may be essential in both inhibiting and promoting skeletal

muscle myogenesis. Evidence for a direct role of NF-κB in the inhibition of satellite cell

differentiation is well documented. Inhibition of NF-κB either via the administration of known

suppressors such as curcumin [213], or genetic manipulation [214], accelerates myogenesis, as

well as myotube formation. Likewise, pharmacological activation of NF-κB inhibits myogenesis

16

in response to severe muscle trauma and in skeletal muscle myopathies [193, 207, 215].

Further, chronic inflammation activates skeletal muscle degradation pathways involved in the

pathology of chronic disease. Chronic activation of NF-κB is observed in several diseased states

and the activation of downstream inflammatory targets are linked to skeletal muscle wasting

and suppressed myogenesis [207, 216-218]. Mechanistically, NF-κB can act through multiple

pathways to block muscle growth and repair. Recent investigations have indicated that NF-κB

inhibits myogenesis via the up regulation of transcriptional factor Ying Yang1 (YY1) which acts

as a repressor of late stage differentiation genes MyoD and MHC [193]. Alternative pathways

that have been explored include the NF-κB regulation of cyclinD1. CyclinD1 is a cyclin-

dependent kinase complex that can cause myoblasts to undergo irreversible growth arrest

[214]. Further, activation of NF-κB by a supraphysiological dose of TNF-α has deleterious

effects on satellite cell signaling [200]. Contrary to these initial findings, numerous in vitro

reports have established a positive association between NF-κB activation and skeletal muscle

myogenesis. Pharmacological inhibition of NF-κB impairs myogenesis in L6 rodent myoblasts

[219, 220]. Both lactacystin [220] and three dimensional clinorotation [219] attenuated

signaling through the classical NF-κB pathway and concomitantly inhibited the expression of

muscle-specific proteins and myoblast differentiation. Whilst our understanding of precise

signaling pathways remains poor, several different mechanisms have been proposed. Briefly,

several mitogenic precursors stimulate or are induced by NF-κB. Growth factors such as PDGF

and VEGF appear to function via an autocrine loop, acting both upstream and downstream of

NF-κB [192, 221]. Further, IGF-II signaling promotes NF-κB DNA binding during C2C12

differentiation via IKKα activation, a key component of the alternative NF-κB signaling pathway

[192]. Correspondingly, NF-κB activity increases the transcription of IGF-BP1 [222, 223] and

IGF-BP2 [224] which is required for terminal myogenic differentiation in the presence of IGF-II.

A number of reports have further identified promyogenic factor p38 MAPK as an activator of

NF-κB signaling through the classical signaling pathway [225-227]. Baeza-Raja et al. [225-227]

established a p38-dependent increase in NF-κB DNA binding activity during myogenic

differentiation in C2C12 cells. It was proposed that NF-κB activation is required for IL-6

production, a cytokine implicated in myogenesis and satellite cell signaling [225]. It is also

plausible to consider a role for TNF-α in this pathway. This cytokine is a known transcriptional

target of NF-κB and at high levels acts as a potent inhibitor of myogenesis in skeletal muscle

myopathies. However, recent in-vitro research discovered low levels of TNF-α promotes

17

myogenesis via signaling through the myogenic activity of p38 MAPK [228], establishing a

conceivable link between NF-κB and TNF-α in skeletal muscle satellite cell activation [229]. This

premise is validated by the role of TNF- α as a positive regulator of myogenesis in cardiotoxin-

injured muscle [230]. Additionally NF-κB induces hypertrophic growth in cardiomyocytes in-

vitro [231]. The discrepancies that exists within the literature suggest that the effects NF-κB

and TNF-α signaling exert on myogenic pathways is likely to be specific to the activating

stimulus, the timing and duration of activation and the precise NF-κB pathway that is activated.

1.5.4 NF- κB and exercise:

Acute exercise triggers a transient rise in various components of the NF-κB signaling pathway

in various cell types [232-236]. However, a precise functional role for NF-κB in contraction-

induced muscle damage remains speculative. Key components of the NF-κB signaling pathway

are up regulated in human peripheral blood lymphocytes and rodent skeletal muscle tissue

immediately post exercise, implicating a potential role for NF-κB in acute muscle damage [232,

234, 235]. Further, this rise appears to peak between 1 and 5 h post exercise, a time point that

coincides with the recruitment and activation of inflammatory neutrophils [232, 235]. Protein

expression of p50/p65 complex and NF-κB DNA binding remains activated at 24 h post-exercise

[232, 234], however a functional role for NF-κB at this time point was not established.

Interestingly Ho et al. [233] observed no change in NF-κB DNA binding activity at 5 h post

exercise in rodent skeletal muscle tissue whilst Ji et al. [234] employed a similar research

model, and identified an increase in NF-κB DNA binding at 4 and 24 h. Collectively these two

experiments may provide an insight into the regulatory patterns of NF-κB following exercise.

Models of TNF-α induced activation of NF-κB and ischemic reperfusion injury, have identified a

biphasic activation pattern of NF-κB in response to cellular stress [216, 237]. The first phase is

transient in nature and appears to peak between 30 min and 3 h post exercise. Activation of a

secondary phase differs between studies, with TNF-α triggering an increase in NF-κB

transactivation function between 24 and 36 h post exposure [216] and ischemic reperfusion

injury causing an increase NF-κB DNA binding between 6 and 16 h [237]. The discrepancy in the

literature is likely to reflect a difference in the stress induced stimulus. This also highlights a

need for further research to examine the time course activation of both the classical and

alternative NF-κB signaling pathways following exercise-induced muscle damage.

18

Understanding the complexity of this pathway and the potential interactions between classical

and alternative NF-κB pathways will provide new insights into the mechanistic link between

exercise-induced inflammation and cellular regeneration pathways.

Remarkably, a paucity of research exists examining the effect of exercise on NF-κB signaling in

human skeletal muscle tissue. Durham et al. [238] conducted an initial trial examining the

effect of an acute resistance exercise bout on the activation of the NF-κB pathway. This

research established a 44% decrease in NF-κB DNA binding activity at 10 min post exercise and

no change at 1 h post [238]. Contrastingly, Tantiwong et al. [236] used a moderate intensity

cycling protocol and observed no change in DNA binding activity immediately post exercise and

a 90% increase at 210 min post. It is likely that the observed disparities between the two trials

may be caused by the differences in exercise protocol or the difference in the time course of

muscle biopsy sampling. Further research from Vella et al. [239] demonstrated a significant

increase in NF-κB (p65) in response to traditional resistance exercise at 2 h post-exercise.

These findings led to the novel exploration for a potential target of NF-κB (p65) in response to

a resistance exercise stimulus and identified NF-κB as a transcriptional regulator of pro-

inflammatory myokines MCP-1, IL-6 and IL-8. EMSA data revealed that NF-κB binding to the

promoter region on all three respective genes increased significantly at 2 h post exercise and

returned to basal levels by 4 h. While these findings establish NF-κB as a key regulator of post-

exercise inflammatory pathways, the regulatory circuitry of the NF-κB pathway, and the

heterogeneity of NF-κB target genes, suggests that our understanding of NF-κB signaling

following exercise is incomplete. Future research needs to consider the activation of both

classical and alternative NF-κB pathways beyond the 4 h time point examined in human

research, and further explore the role of NF-κB in post-exercise inflammatory resolution and

skeletal muscle myogenesis.

1.6 Summary:

The human response to resistance exercise presents a highly complex paradigm. Simplistically,

the outcome of post-exercise inflammation can be regarded as a battle between those

mechanisms that augment the inflammatory response and cause further injury, against those

19

that tend to promote the resolution of inflammation and skeletal muscle repair. The skeletal

muscle response to exercise-induced muscle damage occurs in a well described schematic of

events; degeneration, inflammation and, regeneration. While processes of degeneration and

inflammation can exacerbate symptoms of muscle injury and promote sensations of pain, a

considerable body of literature has established a clear link between signaling pathways in

inflammation and skeletal muscle adaptation. Developing an understanding of the molecular

mechanisms regulating this physiological link is vital in establishing therapeutic treatment

strategies to optimise muscle recovery from exercise-induced tissue injury.

The acute post-exercise inflammatory response is self-resolving in nature and recent works

have identified a series of novel lipid derivatives that play a role in regulating a biologically

active inflammatory resolution program. While this concept remains largely unexplored in

humans post-exercise, initial research demonstrates these lipids are increased in human serum

samples following exercise and may provide a novel mechanism in understanding the link

between inflammation and tissue adaptation [102]. Further, NF-κB is a transcription factor

shown to couple cellular stress with an adaptive or maladaptive response. It has been

implicated in post-exercise inflammation and also demonstrated to be involved in an in-vitro

model of acute inflammatory resolution. The concept that events occurring early in acute

inflammation engage an active cellular resolution program that is intimately necessary for the

activation tissue adaptive pathways, presents a novel and unexplored link between cellular

stress and tissue regeneration that may provide insight into the mechanisms that regulate

post-exercise muscle recovery.

1.7 General hypothesis:

The overall hypothesis of this thesis is that an acute inflammatory resolution pathway exists in

post-exercise inflammation. A biologically active inflammatory resolution pathway provides a

potential mechanism through which cellular events occurring early within an acute

inflammation engage a complex cascade that ultimately leads to the restoration of tissue

homeostasis and skeletal muscle adaptation. NF-κB presents a transcription factor involved in

processes of acute inflammation, inflammatory resolution and myogenesis. Interactions

20

between classical and alternative NF-κB signaling pathways may provide a transcriptional

target that links these cellular processes and plays an integral role in recovery from exercise-

induced muscle damage. This hypothesis will be tested by investigating the specific aims

outlined below.

21

1.8 Specific Aims:

1)

a) To investigate if oral administration of ibuprofen influences skeletal muscle leucocyte

infiltration during the first 24 h after muscle-damaging resistance exercises.

b) To determine if individual changes in leucocyte infiltration are correlated to markers of

muscle damage, including circulating muscle proteins CK and myoglobin, and subjective

markers of muscle soreness.

2) To determine the effect of acute resistance exercise on a novel family of lipid derived

eicosanoids and docosanoids that play a key role in driving a biologically active

inflammatory resolution program.

3) To examine the effect of an acute bout of resistance exercise on regulating members of the

classical and alternative NF-κB signaling pathways in skeletal muscle of young untrained

men.

1.9 Hypothesis:

1. The ingestion of ibuprofen following acute resistance exercise will not influence the

infiltration of leucocytes, however it will attenuate sensations of muscle soreness.

2. Acute resistance exercise will trigger an increase in a novel series of lipid-derived

inflammatory mediators that are involved in regulating an active inflammatory

resolution pathway.

3. Acute resistance exercise will trigger an increase in key parameters of the NF- κB

signaling pathways. The activation of NF-κB will be biphasic in nature, involving the

activation of the classical NF-κB pathway at the onset of inflammation, and the

alternative NF-κB pathway coincident with the resolution of acute inflammation.

22

CHAPTER 2:

Ibuprofen ingestion does not affect markers of post-exercise muscle inflammation.

Vella L, Markworth JF, Paulsen G, Raastad T, Peake JM, Snow RJ, Cameron-Smith D, Russell

AP. Ibuprofen ingestion does not affect markers of post-exercise muscle inflammation.

Frontiers in Exercise Physiology, 2016 Mar 29;7:86. doi: 10.3389/fphys.2016.00086

(Appendix A).

23

2.1 Abstract:

Purpose: Chapter 2 investigated if oral ingestion of ibuprofen influenced leucocyte recruitment

and infiltration following an acute bout of traditional resistance exercise. Methods: Sixteen

male subjects were divided into two groups that received the maximum over-the-counter dose

of ibuprofen (1200 mg d-1) or a similarly administered placebo following lower body resistance

exercise. Muscle biopsies were taken from m.vastus lateralis and blood serum samples were

obtained before and immediately after exercise, and at 3 h and 24 h after exercise. Muscle

cross-sections were stained with antibodies against neutrophils (CD66b and MPO) and

macrophages (CD68). Muscle damage was assessed via creatine kinase and myoglobin in blood

serum samples, and muscle soreness was rated on a ten-point pain scale. Results: The

resistance exercise protocol stimulated a significant increase in the number of CD66b+ and

MPO+ cells when measured at 3 h post exercise. Serum creatine kinase, myoglobin and

subjective muscle soreness all increased post-exercise. Muscle leucocyte infiltration, creatine

kinase, myoglobin and subjective muscle soreness were unaffected by ibuprofen treatment

when compared to placebo. There was also no association between increases in inflammatory

leucocytes and any other marker of cellular muscle damage. Conclusion: Ibuprofen

administration had no effect on the accumulation of neutrophils, markers of muscle damage or

muscle soreness during the first 24 h of post-exercise muscle recovery.

24

2.2 Introduction:

Unaccustomed resistance exercise often results in tissue damage and inflammation, leading to

delayed onset muscle soreness (DOMS) and a consequent reduction in force production [31,

240]. Animal models and in vitro studies have identified that local and systemic inflammation

exerts a regulatory influence during the different phases of muscle recovery, including

myofibrillar disruption, cellular necrosis, satellite cell activation, maturation and subsequent

regeneration and adaptation [17, 24]. Thus, post-exercise inflammation is intimately necessary

and a key feature of the normal process of tissue regeneration and adaptation following acute

muscle damage. However, excessive inflammation is considered a potential cause of prolonged

post-exercise muscle soreness and may have a negative effect on muscle recovery [24, 25];

consequently strategies to reduce or counteract inflammation are commonly implemented to

aid in improving muscle recovery after exercise [103].

Non-steroidal anti-inflammatory drugs (NSAIDs) are commonly used as a treatment strategy in

exercise and sports medicine to assist with recovery from exercise-induced inflammation,

particularly following soft-tissue injury. NSAIDs inhibit the cyclooxygenase (COX-1 and 2)

enzymes and consequently the formation of prostanoids (prostaglandins, prostacyclins and

thromboxanes) that play a diverse roll in acute inflammation [102]. Prostanoids stimulate an

acute inflammatory process by controlling local blood flow, vascular permeability, leucocyte

infiltration, and triggering sensations of pain [102, 103]. In animal models of acute muscle

damage, treatment with NSAIDs blunts the infiltration of leucocytes into muscle tissue [104,

159] and causes a reduction in creatine kinase (CK) [106]. Consequently, NSAIDs can also

inhibit myofiber regeneration, satellite cell proliferation and differentiation, and overload-

induced muscle hypertrophy [104-106]. These findings provide preliminary evidence that

NSAIDs compromise the physiological link between processes of acute muscle damage,

inflammation and cellular regeneration.

Research into the effects of NSAIDs on exercise-induced muscle damage and inflammation has

produced equivocal findings. In exercise models, NSAID administration has been shown to

attenuate post-exercise DOMS in some [93, 140, 241], but not all studies [87, 107, 151, 152].

25

While a precise cellular mechanism for an analgesic effect of NSAIDs remains unclear, it has

been largely ascribed to their effect on prostaglandin synthesis and the capacity of NSAIDs to

interfere with aspects of inflammatory cell function [160, 242]. Previous research has

demonstrated that oral consumption of both ibuprofen, a non-selective NSAID and

acetaminophen, an analgesic also known as paracetamol, had no effect on macrophage

infiltration at 24 h following an eccentric exercise protocol [160]. Similarly treatment with

naproxen, another non-selective NSAID, had no effect on the infiltration of leucocyte common

antigen positive cells following a unilateral, isotonic resistance exercise protocol [147].

Interestingly, Paulsen et al., (2010) suggested a blunting effect of the COX-2 specific celecoxib

following maximum eccentric muscle contractions [93]. This research identified a tendency for

higher monocyte/macrophage numbers in subjects within the placebo group, and those

subjects who were identified as ‘high-responders’ to the exercise protocol based on the

number of inflammatory leucocytes [93]. Although this is not a particularly robust finding, it

has led to the hypothesis that NSAIDs may influence leucocyte infiltration in skeletal muscle

when a sufficiently strong and early inflammatory reaction is present [93]. This hypothesis

would suggest that the intensity of the exercise stimulus and the consequent muscle-damage

response would be largely influential in determining the effect of a pharmacologically based

anti-inflammatory intervention.

Conflicting findings also exist with regard to the effect of NSAIDs on the regenerative capacity

of skeletal muscle following exercise-induced muscle damage. The non-selective COX inhibitor

ibuprofen blunted skeletal muscle protein synthesis [107] while local intramuscular infusion of

indomethacin [243] and the oral administration of the COX-2 selective NSAID celecoxib [93,

130] had no such effect. Similarly, treatment with indomethacin inhibited post-exercise

satellite cell proliferation [87]. Recent work from our group demonstrated that treatment with

ibuprofen inhibited early translational signalling responses involved in post-exercise muscle

hypertrophy [161]. A clear mechanistic pathway for NSAIDs to influence the physiological link

between post-exercise inflammation and skeletal muscle regeneration remains elusive.

26

These discrepancies in the research to date are likely due to differences in the exercise

protocol (concentric vs. eccentric muscle contractions), the training status of subjects, the

timing of muscle biopsies, and the type of NSAID, the dosage administered, and method of

administration. Further research is required to determine whether NSAID administration

affects the infiltration of leucocyte populations following exercise-induced muscle damage and

how this influences post-exercise adaptive pathways. The aim of the present study was to

investigate if oral administration of the NSAID ibuprofen influenced skeletal muscle leucocyte

infiltration during the first 24 h after muscle-damaging resistance exercises. A further aim was

to explore how any changes in leucocyte infiltration were related to markers of muscle

damage, including circulating muscle proteins CK and myoglobin, and subjective markers of

muscle soreness. It was hypothesized that the ingestion of ibuprofen following acute

resistance exercise would not influence the infiltration of leucocytes, however it would

attenuate sensations of DOMS.

27

2.3 Methods:

2.3.1 Participants:

Sixteen healthy male subjects were recruited to participate in the study (Table 1) [102].

Exclusion criteria included participation in a lower body resistance exercise program within the

last 6 months to ensure a muscle damage response from the exercise stimulus, and/or

previous chronic treatments with anti-inflammatory medication. Participants also completed a

medical screening to identify any potential risk factors for them to perform strenuous physical

activity.

Table 2.1: Subject characteristics and strength testing data. Values are mean values ± SEM. No

significant differences were observed between the two groups.

Characteristics Strength (1RM)

Age (y) Height

(m)

Body mass

(kg)

BMI Squat (kg) Leg Press

(kg)

Leg

Extension

(kg)

PLA 23.9

± 1.3

1.89

± 0.1

86.9

± 4.5

24.5

± 1.2

94.9

± 5.4

237

± 17

236

± 18

IBU 23.0

± 0.5

1.89

± 0.1

89.1

± 4.4

24.8

± 0.8

91.9

± 6.0

240

± 15

196

± 22

PLA, placebo; IBU, Ibuprofen; BMI, body mass index

2.3.2 Ethics approval:

All procedures involved in this study were approved by the Deakin University Human Research

Ethics Committee (DUHREC 2010-019) and muscle sampling procedures were performed in

accordance with the Helsinki declaration. Each participant was provided with written and oral

details of the nature and requirements of the study and provided written consent to

participate.

28

2.3.3 Familiarization:

Each participant completed a familiarization and strength testing session at least 7 days prior

to completing the exercise protocol. Briefly, subjects performed repetition maximum testing

for the Smith machine-assisted squat, the leg press and the leg extension to determine their

experimental exercise load (80% of a 1 repetition maximum (1RM)). The maximum weight the

subject could lift for 5-8 reps was determined, and these data were entered into the Brzycki

equation to predict 1RM [1 RM = 100 X load rep/(102.78 – 2.78 X reps completed] [244].

Subjects were asked to abstain from any further activity until the completion of the trial.

2.3.4 Experimental Procedures:

The participants reported to the laboratory on the morning of the trial in an overnight fasted

state. They were asked to abstain from caffeine, tobacco and alcohol for the 24 h preceding

the trial. Participants rested in a supine position for 30 min, following which the first muscle

biopsy sample was taken. Each participant then completed a 10-min warm-up protocol

comprised of 5 min of low intensity cycling on a stationary bike, and one low-intensity set of

each exercise at a weight of each subject’s own choice within the range of 30-50% 1RM. The

resistance exercise session consisted of 3 sets of 8-10 repetitions performed on a Smith

machine assisted squat, a 45-degree leg press and a leg extension at 80% of a predicted 1RM.

The exercises were performed as a circuit with 1 min rest permitted between exercises and 3

min rest between sets. This protocol has been used previously and has been a sufficient

stimulus to activate inflammatory signalling pathways [239] and was implemented to replicate

a commonly used exercise routine. After exercise, the subjects rested while subsequent muscle

biopsy samples were collected. After the 3 h biopsy, participants were provided a standardized

meal and could go home. The following morning, participants reported to the laboratory in an

over-night fasted state for a 24 h muscle biopsy and blood sample.

2.3.5 Standardized meals:

Standardized meals were provided to participants on the night before the trial, (carbohydrate

57%, fat 22%, protein 21%), immediately following the 3 h muscle biopsy (carbohydrate 71%,

fat 13%, protein 16%), and in the evening (carbohydrate 64%, fat 27%, protein 18%) on the day

29

of the exercise trial. All participants received the same meal and individualised energy content

based on body mass was not provided. Participants were permitted to drink water ad libitum

and were asked to report if they could not finish their allocated meals. This nutrition plan was

included to ensure that each participant received the same relative percentage of macro- and

micro-nutrients.

2.3.6 NSAID Administration:

Prior to exercise, participants were randomly assigned in a double-blind method, to consume

either the maximum recommended dose of ibuprofen (IBU, n = 8) or a placebo control (gelatin

capsules identical in appearance containing powdered sugar in place of ibuprofen) (PLA, n = 8).

The IBU group consumed three doses of 400 mg of ibuprofen throughout the trial day. The first

dose was administered immediately prior to the first muscle biopsy sample. Participants were

instructed to consume the following two doses at 6 h and 12 h following the exercise protocol.

Follow-up phone calls from the research team ensured compliance. To ensure this dosing

structure was appropriate to maintain biologically active levels of ibuprofen, ibuprofen

concentration was measured in blood serum samples and these data are reported elsewhere

[102].

2.3.7 Sample collection:

Venous blood samples were drawn prior to exercise, within 5 min post exercise (here-after

referred to as 0 h post-exercise), and at 1, 2, 3 and 24 h post-exercise. Blood samples were

drawn through an indwelling catheter into VACUETTE serum tubes (Greiner,Bio-One,

Stonehouse UK). Whole blood was allowed to clot at room temperature. Samples were then

centrifuged at 1,000 g for 10 min. The serum layer was collected and stored at -80°C for

subsequent analysis.

Muscle biopsy samples were obtained from the vastus lateralis muscle under local anaesthesia

(Xylocaine 1%) using a percutaneous needle biopsy technique modified to include suction

[245]. To minimize interference from the biopsy procedure, samples prior to exercise and at 24

30

h post exercise were taken from the same leg, while samples taken at 0 and 3 h post-exercise

were taken from the contralateral leg. Samples obtained within the same leg were taken at

least 5cm from the previous incision site. A section of excised tissue was immersed in Tissue-

Tek and stored at -80°C until further analysis. We have previously reported that this technique

is effective for minimizing the risk of any inflammation arising from the biopsy procedure

confounding exercise-induced inflammation [239]. This research showed no effect of the

biopsy protocol on mRNA expression of acute phase inflammatory cytokines, however did not

directly investigate a potential effect on leucocyte infiltration or markers of cellular muscle

damage. The absence of a non-exercise control group is a limitation of the present study.

2.3.8 Immunohistochemistry:

Cross sections (8 μm) of muscle tissue were cut using a microtome at -20 ⁰C (CM3050, Leica,

Germany) and mounted on microscope slides (Superfrost Plus, Thermo Scientific, MA, USA),

air-dried and stored at -80 °C. Each subject’s muscle sections from all time points were

mounted on the same microscope slide. Serial muscle cross-sections were stained with three

different leucocyte antibodies, MPO (#0398, DakoCytomation, Glostrup, Denmark; dilution

1:2000), CD66b (#CLB-B13.9, Sanquin Reagents, Amsterdam, The Netherlands; dilution 1:500)

and CD68 (#EBM-11, DakoCytomation, Glostrup, Denmark; dilution 1:300). Furthermore, the

sections were also stained with dystrophin antibody (ab15277, Abcam, Cambridge, UK) for

visualizing borders of muscle fibers

.

Sections were fixed in 4 % paraformaldehyde solution in a staining jar for 5 min at room

temperature and rinsed twice for 10 min in phosphate buffered saline (PBS) containing 0.5 %

Tween 20 (Sigma-Aldrich). The microscope slides were moved into a humidified chamber and

non-specific binding sites were blocked with 1% bovine serum albumin (BSA) on the section for

45 min at room temperature. Sections were incubated overnight with leucocyte and

dystrophin antibodies diluted in 1% BSA at 4°C. After overnight incubation, the slides were

washed three times for 10 min in PBS-Tween in a staining jar. Slides were moved back to the

humidified chamber and sections were incubated for 45 min with secondary antibodies diluted

1:200 in 1% BSA at room temperature. Alexa Fluor® 594 F(ab’)2 fragment of goat anti-rabbit

31

and Alexa Fluor®488 anti-mouse IgG (Invitrogen, Eugene, Oregon, USA) were used as a

secondary antibodies. The fluorochrome-stained sections were washed three times for 10 min

in PBS-Tween. After the last washing in PBS-Tween, the sections were mounted on ProLong®

Gold Antifade reagent with DAPI (Invitrogen, Eugene, Orgeon, USA).

Muscle sections were visualized using a high-resolution camera (DP72, Olympus, Japan)

mounted on a microscope (BX61, Olympus, Japan) with a fluorescence light source (X-Cite

120PCQ, EXFO, Canada). The number of MPO, CD66b and CD68 positive cells (as well as the

total number of muscle fibres from the area included) was counted. Data was calculated as

percentage of MPO, CD66b or CD68 positively stained cells per 100 skeletal muscle fibres.

Areas of sections that contained freeze damage or were folded due to the cutting procedure

were not included in the analysis.

2.3.9 Biochemical assays:

Serum creatine kinase activity in pre- and post-exercise blood samples was analyzed using an

enzymatic assay (CK-NAC kit, CDT14010, Thermo-Fisher Scientific Clinical Diagnostics, Sydney,

Australia) and an automated clinical analyser (Cobas Mira, Roche Diagnostics, Germany).

Serum myoglobin concentration was also measured in these samples using an immunoassay

(Roche Diagnostics, Germany) and an automated clinical analyzer (Cobas E411, Roche

Diagnostics, Germany). The intra-assay coefficient of variation was 10.4% for creatine kinase

and 1.7% for myoglobin.

2.3.10 Muscle soreness assessment:

Upon arrival at the laboratory and prior to the first muscle biopsy procedure subjects were

asked to rate their subjective muscle soreness on a 0-10 visual analogue scale. The subjects

were instructed to contract, stretch and palpate the quadriceps muscle to assess general

muscle soreness. In both instances 0 was considered to represent no pain and correspondingly

a rating of 10 was considered to represent intense pain.

32

2.3.11 Statistics:

Data are expressed as means ± SEM. Prior to analysis the data displaying a lack of normality

were log transformed to stabilise variance. Data were analysed using a two-way ANOVA with

repeated measures for time. The sphericity adjustment was checked, and if required, a

Greenhouse-Geisser epsilon correction was applied. Where no significant effect for treatment

was observed, we explored pair-wise comparisons between individual time-points using the

Least Significant Difference (LSD) of means. Bivariate relationships were examined with a

Spearman rank correlation test. Confidence intervals were calculated at 90% using a

bootstrapping technique for non-normally distributed data. Statistical analyses were

performed using GenStat for Windows 16th Edition (VSN International, Hemel Hempstead, UK).

Statistical significance was set at P < 0.05.

33

2.4 Results:

2.4.1 Muscle Soreness:

Muscle soreness showed a main effect for time (p < 0.01), suggesting that the exercise protocol

was sufficient to induce a DOMS response. No effect of the ibuprofen treatment was observed

(Figure 2.1).

Figure 2.1: Subjective rating for delayed onset muscle soreness (DOMS) (B). Values depicted are

mean values ± SEM. * denotes statistical significance from pre-exercise values (p<0.05). White bars

= PLA group; black bars = IBU group. This figure has been adapted from Vella et al. (2014) [246].

2.4.2 Immunohistochemistry:

The number of MPO+ cells per 100 myofibres analysed showed a main effect for time (P <

0.01). No main effect for treatment (P = 0.250) or time x treatment (P = 0.709) was observed

(Figure 2.2A). LSD pairwise comparisons indicated a significant increase in MPO+ cells at all

sampling time points post-exercise. The greatest increase in MPO+ cells was observed at 3 h

post-exercise (Figure 2.2B). Similarly, the number of CD66b+ cells per 100 myofibres showed a

main effect for time (P < 0.05), with no main effect for treatment (P = 0.149) or time x

treatment interaction (P = 0.907) (Figure 2.2C). LSD pairwise comparisons indicated a

significant increase in the number of CD66b+ cells at 3 h post exercise (Figure 2D). The number

of CD68+ cells per 100 myofibres analysed showed no effect for time (P = 0.061), treatment (P

= 0.530) or time x treatment interaction (P = 0.688) (Figure 2.2D). MPO+, CD66b+, and CD68+

cells were observed within the endomysium and the perimysium, with no cellular infiltration

occurring at any time point (Figure 2.2E). Representative images for each cell surface marker

are presented in Figure 3.

34

Figure 2.2: Immunohistochemistry data for muscle cells staining positive for myeloperoxidase

(MPO), showing group specific data (A) and collapsed data (B), CD66b showing group specific data

(C) and collapsed data (D), and CD68 showing group data only (E). Data represent the mean number

of positively stained cells per 100 fibres analysed ± SEM. ** denotes statistical significance from

pre-exercise values (p<0.01). * denotes statistical significance from pre-exercise values (p<0.05). ^

denotes statistical significance from 0 h post-exercise (p<0.05). White bars =PLA group; black bars =

IBU group.

35

Figure 2.3: Immunohistochemical analysis of skeletal muscle samples following a bout of resistance

exercise. Sections were probed with antibodies raised against leucocyte cell surface markers (green;

Figure 3A– MPO, Figure 3B – CD66b, Figure 3C – CD68) and dystrophin (red), while DAPI was used

to stain nuclei (blue). Scale bars = 50µm.

2.4.3 Serum proteins:

Serum CK activity demonstrated a main effect for time (P < 0.01) with no effect for treatment

(P = 0.843) or time x treatment interaction (P = 0.494) (Figure 2.4A). LSD comparisons revealed

that serum CK increased from pre-exercise values at 2 h post-exercise and peaked at 24 h post-

exercise (Figure 2.4B). Similarly, serum myoglobin concentration showed a main effect for time

(P < 0.01) with no effect for treatment (P = 0.710) or time x treatment interaction (P = 0.666)

(Figure 2.4C). LSD comparisons demonstrated that the biggest increase in serum myoglobin

concentrations occurred at 1 h post-exercise and remained significantly elevated up to 3 h post

exercise (Figure 2.4D).

36

Figure 2.4: Blood derived proteins creatine kinase (A and B) and myoglobin (C and D). Values

depicted are mean values ± SEM. Figures A and C represent data divided into two treatment

groups; figures B and D represent collapsed data. *denotes statistical significance from pre-exercise

values (p<0.05). ** denotes statistical significance from pre-exercise values (p<0.01). ^ denotes

statistical significance from 1, 2 and 3 h post exercise (p<0.05). White bars = PLA; black bars = IBU.

2.4.4 Correlation analysis:

In general, large inter-individual differences were seen in the appearance of all of the leucocyte

cell types. Results from the Spearman rank correlation test showed no correlation between the

histochemical appearance of inflammatory leucocytes, biological markers of muscle damage

and subjective markers of muscle soreness (Table 2). Anecdotally, three subjects were

37

identified as the highest responders in the appearance of cell surface markers applicable for

localization of neutrophils (one subject within the placebo group, and two subjects within the

ibuprofen group). However, these subjects showed no significant increase in CK, myoglobin or

subjective markers of muscle damage when compared with the other subjects. Interestingly,

subject 5 recorded the highest number of CD66b+ cells and MPO+ cells, whereas he presented

with the lowest subjective rating for DOMS.

Table 2.2: Spearman rank correlation analysis. Values depicted are the correlation coefficient

with 90% confidence intervals in parenthesis. Statistical significance was set at p < 0.05. No

statistically significant correlations were observed.

CK MYO CD68 CD66b MPO DOMS

CK - 0.59

(-0.01 - 0.91) -0.46

(-0.88 - 0.8) 0.15

(-0.42 - 0.61) 0.36

(-0.14 - 0.73) 0.01

(-0.60 - 0.62)

MYO - - 0.01

(-0.55 - 0.52) 0.14

(-0.42 - 0.69) 0.35

(-0.21 - 0.67) -0.33

(-0.74 - 0.24)

CD68 - - - 0.05

(-0.46 - 0.56) -0.27

(-0.75 - 0.33) -0.37

(-0.82 - 0.25)

CD66b - - - - 0.41

(-0.13 - 0.84) -0.37

(-0.78 - 0.16)

MPO - - - - - 0.08

(-0.49 – 0.56)

DOMS - - - - - -

CK, creatine kinase; MYO, myoglobin; MPO, Myeloperoxidase; DOMS, delayed-onset muscle

soreness.

38

2.5 Discussion:

The main finding of this study was that ibuprofen, a non-selective COX inhibitor, had no effect

on the histological detection of leukocytes following an acute bout of traditional resistance

exercise. Serum CK and myoglobin, and muscle soreness also did not change in response to

ibuprofen. Furthermore, no correlation was observed between the accumulation of

inflammatory leucocytes, increases in CK or myoglobin, and sensations of DOMS. These

observations suggest that the intramuscular infiltration of inflammatory white blood cells, or

an increase in serum levels of intramuscular proteins, are not predictive of post-exercise

muscle soreness.

The current study focused on the acute local inflammatory response to exercise-induced

muscle damage. Our protocol was effective in inducing muscle soreness as shown in Figure 2.1.

An increase in the histological appearance of CD66b+ and MPO+ cells occurred at 3 h post

exercise, while no effect of the NSAID treatment was observed. CD66b is a highly specific cell

surface marker for detecting human neutrophils, and this is the first paper to show a small but

significant increase in the intramuscular number of CD66b+ cells following acute resistance

exercise. Paulsen et al. (2013) were unable to identify any change in the number of CD66b+

cells in the elbow flexors at 1 h, 2, 4 or 7 days following 70 maximal eccentric contractions [97].

The discrepancy between the two trials may be the result of a combination of methodological

differences, such as the timing of muscle biopsy samples, the mode of exercise and the muscle

group sampled. A small but significant increase in the number of MPO+ cells at all sampled time

points was also observed, with a peak in expression at 3 h post exercise. MPO has typically

been used to detect changes in neutrophils, and an increased number of MPO+ cells have been

reported following high-force eccentric muscle contractions [98, 247]. However, MPO is also

expressed on the lysosomes of monocytes and has been detected on basophils and

eosinophils, perhaps providing a reason why there were a higher number of MPO+ cells when

compared to CD66b+ cells detected in the present study [97].

No change was identified in the number of CD68+ cells. CD68 cell counts are widely used as a

marker of monocyte/macrophage infiltration. However, CD68 may also be expressed on other

39

cell types including satellite cells and fibroblasts [97]. Other researchers have been able to

identify a change in CD68+ cells following exercise-induced muscle damage [98, 160, 247]. Each

of these studies have used a high-force maximal eccentric muscle contraction protocol, which

may suggest that the detection of CD68+ cells is dependent on the extent of skeletal muscle

damage.

Previous research exploring the effect on NSAIDs on post-exercise inflammation has produced

equivocal findings. Earlier research from in-vivo rodent trials demonstrated a blunted

inflammatory response to eccentric muscle contractions following the administration of both

selective and non-selective NSAIDs [104, 105, 248, 249]. However, these findings are yet to be

supported in human trials [140, 147, 160]. Petersen et al. (2003) found no effect of oral

ibuprofen administration on the appearance of CD68+ cells 24 h following 100 eccentric muscle

contractions [160]. Similarly, Tokmakidis showed no effect of ibuprofen on circulating white

blood cells 24 h following an eccentric exercise protocol [140]. Paulsen et al. (2010) proposed

the concept of an inflammatory threshold, suggesting that NSAIDs may only influence post-

exercise inflammation in response to a sufficiently strong inflammatory stimulus [93]. It was

suggested that the absence of an intramuscular prostaglandin response to exercise, specifically

PGE2, could explain the lack of an NSAID effect on leucocyte recruitment [93, 132].

Interestingly, our group recently reported an increase in serum PGE2 in samples that were

obtained during the same exercise trials as those currently reported [102]. This change in

serum PGE2 was blunted by the administration of ibuprofen and suggests that in the presence

of a blunted prostaglandin response to exercise, NSAID treatment had no effect on leucocyte

recruitment within skeletal muscle. Although this finding does not refute the possibility of an

inflammatory threshold, based on our previous findings [102], it is reasonable to suggest that

the presence of an exercise-induced serum prostaglandin response is not the required

underlying mechanism for neutrophil infiltration of muscle during the early hours of post-

exercise recovery.

In accordance with previous studies, resistance exercise resulted in an increase in serum CK

and myoglobin, and subjective muscle soreness [250]. Although large inter-individual variations

40

in the human response to exercise can make it difficult to establish causal relationships, there

was no relationship between the appearance of leucocytes and blood-derived markers of

muscle damage or sensations of muscle soreness. Interestingly, the subject with the highest

number of CD66b+ and MPO+ cells presented with the lowest subjective rating of DOMS. These

findings provide support for the assumption that leucocyte recruitment and inflammation do

not consistently cause DOMS.

From animal studies, it appears that cellular events in post-exercise inflammation, occur

concomitantly with the onset of DOMS [24, 240]. The mechanistic link has been proposed to be

a leucocyte-mediated release of cytotoxic substances and reactive oxygen species as a bi-

product of phagocytosis [251]. Furthermore, the prostaglandin response to exercise has been

shown to occur concurrently to the onset of DOMS, and is known to influence both the

recruitment of inflammatory leucocytes and neural afferents to pain [102]. However, in line

with the current findings, the accumulation of inflammatory leucocytes in human skeletal

muscle tissue during exercise recovery appears to be both spatially and temporally out of

phase with the development of DOMS [93]. Malm et al. [252] explored the concept that the

activation of local leucocytes present in the epimysium may be involved in the regulation of

DOMS. This hypothesis has since received support from both rodent [253] and human trials

[61, 254] and presents a promising area for future research.

2.6 Conclusion:

This is the first study to demonstrate that ibuprofen administration has no effect on the

histological appearance of inflammatory white blood cells following an acute bout of

traditional resistance exercise. Ibuprofen administration had no effect on blood markers of

muscle damage or subjective muscle soreness, and no significant correlations between

leucocyte numbers and post-exercise muscle damage or soreness was observed. Future

research needs to explore the possibility of an inflammatory threshold above which NSAID

administration influences post-exercise inflammation. The underlying cause of post-exercise

muscle soreness also remains largely unknown. Future investigations are needed to determine

the role that inflammation plays in exercise-induced DOMS.

41

2.7 Acknowledgements:

The authors would like to acknowledge to assistance of Associate Professor John Reynolds for

his contributions in the statistical analysis of the dataset. We further thank Dr. Andrew

Garnham (Deakin University) for all muscle biopsy procedures, and the participants for

volunteering their time to be involved in the trial.

2.8 Disclosures:

This research was conducted in the absence of any commercial or financial relationships that

could be construed as a potential conflict of interest.

42

CHAPTER 3:

Intramuscular inflammatory lipid profile response to an acute bout of resistance exercise in

humans.

Vella L, Markworth JF, Maddipati KR, Cameron-Smith D, Russell AP. Intramuscular

inflammatory lipid profile in response to an acute bout of resistance exercise in humans.

Submitted for review to Frontiers in Physiology: Exercise Physiology on 20/11/2016.

43

3.1 Abstract:

Purpose: Classic eicosanoid species including prostaglandins (PG) and leukotrienes (LT) play

essential roles in initiating and regulating acute inflammation. Recent identification of novel

classes of bioactive lipid mediators derived from long-chain (LC) polyunsaturated fatty acids

(PUFA), collectively termed specialized resolving mediators (SPM) has led to the proposal of an

active biochemical pathway for resolution of the acute inflammatory response. Chapter 3

investigated the human intramuscular lipid mediator profile at rest and in response to an acute

bout of unaccustomed resistance exercise. Methods: Fourteen male subjects completed a bout

of lower body resistance exercise. Muscle biopsy samples were taken from the m.vastus

lateralis before and at 2, 4 and 24 h post-exercise. Intramuscular lipid mediator composition

was analysed via liquid chromatography–mass spectrometry (LC-MS)-based targeted

lipidomics. Results: A total of 129 unique lipid mediators were reliably identified in human

skeletal muscle tissue. Post-exercise recovery was characterized by increased levels of COX-

derived thromboxane (TXB2 and 12(S)-HHTrE) and prostaglandin metabolites (2,3-dinor PGE1

and 15d-D12,14-PGJ3), LOX derived leukotrienes (LTB4 and 20-COOH LTB4),

hydroxyeicosatetraenoic acids (5-HETE, 12-HETE, tetranor 12-HETE and 15-HETE),

monohydroxylated- docosahexaenoate (HDoHE) species (4-HDoHE, 7-HDoHE and 8-HDoHE),

and CYP derived epoxyeicosatrienoic acids (5,6-EpETrE) / vicinal diol dihydroxyeicosatrienoic

acids (11,12-DiHETrE and 14,15-DiHETrE). Conclusion: This research utilised a targeted

lipidomics approach to profile the human skeletal muscle lipid mediator response to an acute

bout of resistance exercise. The profile suggests that the activation of classical pro-

inflammatory eicosanoids such as PG and LT species, as well as novel lipid intermediates in

SPM biosynthesis derived from both LOX and CYP pathways, form a highly complex self-

resolving model of acute inflammation following an acute bout of resistance exercise.

44

3.2 Introduction:

Skeletal muscle is a remarkably heterogeneous tissue with the capacity to adapt and respond

to external stress. It is well established that intense resistance exercise can lead to

improvements in muscle strength through changes in muscle fiber type, myofibrillar

hypertrophy and neuromuscular mechanisms [1]. However, unaccustomed exercise, especially

when comprising a large eccentric component, can cause skeletal muscle injury and initiate an

acute inflammatory response [24, 31, 240]. Experimental models targeted at manipulating the

post-exercise inflammatory response have identified that exercise-induced inflammation is a

key regulatory feature in the normal process of tissue regeneration and adaptation following

acute muscle damage [103, 161, 255]. This suggests that molecular signaling events occurring

early during acute inflammation play an active role in promoting the restoration of normal

tissue function and promote skeletal muscle adaptation following an exercise stimulus.

The humoral and local muscular changes that occur during exercise-induced inflammation

closely resemble that of an acute phase response to cellular stress. As observed in chapter 2, a

resistance exercise stimulus triggers the production of pro-inflammatory signaling molecules,

establishing a chemotactic gradient and the diapedesis as well as the infiltration of

inflammatory leukocytes [85, 246]. These chemoattractants consist of lipid-derived mediators

such as leukotrienes (LTs) and prostaglandins (PGs), as well as protein mediators, including

cytokines and chemokines [168]. The usual outcome of an acute inflammatory response is its

successful resolution and repair of damaged tissue [174]. Traditionally, the resolution of

inflammation was thought to be a passive process involving the dilution and catabolism of pro-

inflammatory mediators leading to exodus of leukocytes from the site of muscle damage.

However, with the discovery of novel classes of lipid-derived mediators, the resolution of acute

inflammation has evolved to be seen as an active and finely controlled biochemical and

metabolic process that may provide a critical link between cellular stress and tissue

regeneration/adaptation [185, 186].

Lipid mediators are biosynthesized endogenously from essential omega-6 (n-6) and omega-3

(n-3) polyunsaturated fatty acids (PUFA) and are involved in a wide range of physiological and

45

pathophysiological processes [186]. The majority of research has focused on classic

prostaglandins (synthesized via cyclooxygenase (COX) enzymes, COX-1 and COX-2) and

leukotrienes (synthesized via the lipoxygenase (LOX) enzyme, 5-LOX), which are derived from

the n-6 PUFA arachidonic acid (AA). These lipids play a diverse role in stimulating acute

inflammation by controlling local blood flow, vascular permeability, cytokine production,

leucocyte chemotaxis and pain [102]. More recently, a second class of eicosanoids also

generated from AA, termed lipoxins (LX) [172, 175, 176], as well as newly identified

eicosapentaenoic (EPA) (E-Series) and docosahexaenoic acid (DHA) (D-Series) derived resolvins

(Rv), protectins (PD) [166, 169-171, 177, 178], and maresins (MaR) [168, 173] have been shown

to play pro-resolution functions following acute inflammation. These novel lipid mediators,

collectively termed specialized pro-resolving mediators (SPMs), act to block acute

inflammatory signals by inhibiting pro-inflammatory cytokine production and subsequent

neutrophil chemotaxis [173, 182]. They simultaneously promote the nonphlogistic infiltration

of blood monocytes/macrophages and stimulate tissue macrophages to phagocytize and clear

apoptotic neutrophils whilst promoting wound healing [175, 181].

SPMs are formed during inflammatory transcellular interactions, involving the sequential

actions of two or more cell types expressing the required LOX and/or COX enzymes in a

compartmentalized manner. During the time-course of inflammation, cell-cell interactions

between platelets, leucocytes, the vasculature and resident tissue cells facilitates the

transcellular biosynthesis of unique SPMs [101]. The temporal regulation of these lipids is

therefore specific to the tissue type and the inciting inflammatory stimulus[185, 186]. For

example, LX biosynthesis involves cellular interactions between 5-LOX expressing neutrophils

with 12-LOX expressing platelets or 15-LOX expressing M2 monocytes [184]. Recent findings

from Markworth et al. (2013) demonstrated that pro-resolving lipid mediators, including

lipoxins, resolvins and protectins, are elevated in human blood serum samples following an

acute bout of resistance exercise [102]. The present study used the same targeted lipidomics

approach to characterize the local time-course changes of eicosanoid and docosanoid species

in human skeletal muscle tissue following an acute bout of resistance exercise. The primary

aim was to identify which species of lipids are present within skeletal muscle tissue, and hence

may be locally generated and acting, following an acute bout of resistance exercise. It was

46

hypothesized that there would be a rapid increase in pro-inflammatory prostaglandin and

leukotriene biosynthesis, followed by the activation of SPMs at the onset of inflammation

resolution. Identification of the lipid mediator profile of skeletal muscle and ability of exercise

stress to modulate intramuscular bioactive lipids will help to contribute to the understanding

of a biologically active inflammatory resolution pathway that may be essential to muscle

recovery and adaptation following an inflammatory event.

47

3.3 Methods:

3.3.1 Subjects:

As previously described [256], fourteen healthy men aged 18-25 years were recruited to

participate in the acute exercise study. A subset of 12 male participants, for which sufficient

muscle biopsy tissue remained, were included in the analysis performed here. Exclusion criteria

included participation in regular resistance exercise within one year prior to commencing the

study, and/or the consumption of any nutritional or purported muscle building supplements.

Each participant also completed a medical history questionnaire to identify any potential risk

factors that would prevent the subjects from completing strenuous exercise.

3.3.2 Ethics approval:

Each participant was provided with a written and oral explanation of the nature of the study

and potential risks of the experimental procedures before providing written consent to

participate. All procedures involved in the study were formally approved by the Deakin

University Human Research Ethics Committee (DUHREC 2004-017) and muscle biopsy

procedures were performed in order with Helsinki declaration.

3.3.3 Familiarization:

For the 24 h preceding exercise, subjects consumed a standardised diet (20% fat, 14% protein,

and 66% carbohydrate) [257]. At least seven days prior to the trial day, each subject completed

a familiarization session on the Cybex NORM dynamometer (Cybex International Inc. UK). The

session involved performing isokinetic maximal voluntary contractions (iMVC) during

concentric and eccentric knee extension exercise. Maximal force production measured as peak

torque (N.m) was determined at 60°/s over 12 maximal concentric and eccentric contractions.

Subjects were provided with verbal encouragement throughout the test to ensure maximal

effort.

48

3.3.4 Experimental design:

On the morning of the trial, subjects reported to the laboratory in an overnight fasted state

having abstained from alcohol, caffeine and tobacco for the previous 24 h. Participants rested

in a supine position for 30 min, following which a resting muscle biopsy sample was collected.

Each participant then completed an acute bout of concentric and eccentric isokinetic unilateral

knee extension exercise on the Cybex NORM dynamometer. Subjects completed three sets of

12 maximal voluntary repetitions at a constant speed of 60°/s with 2 mins of rest between

each set. In a previous research project, this exercise stimulus was shown to trigger an increase

in pro-inflammatory gene expression, STAT-3 phosphorylation and nuclear translocation, and

STAT-3 target gene expression [257]. Subjects were instructed to contract maximally during

each repetition and were provided with verbal encouragement throughout each set. Further

muscle biopsy samples were obtained from the exercised leg at 2 and 4 h after completion of

the exercise protocol. The following morning, subjects reported to the laboratory again in an

overnight fasted state for a final follow up 24 h post-exercise muscle biopsy sample.

3.3.5 Muscle biopsy procedure:

Muscle biopsy samples were obtained from vastus lateralis under local anaesthesia (Xylocaine

1%) using a percutaneous needle biopsy technique modified to include suction [245]. A section

of excised tissue was rapidly snap frozen in liquid nitrogen and stored at -80°C for further

analysis. Repeat muscle biopsy samples were collected from the same leg through separate

incisions separated byat least 2 cm from the previous biopsy site to minimize the risk of any

localized inflammation arising from the biopsy procedure confounding exercise-induced

inflammation.

3.3.6 Liquid chromatography-mass spectrometry:

Muscle biopsy samples were weighed and homogenized in 1 ml phosphate buffered saline (50

mM phosphate containing 0.9% sodium chloride, pH 7.4) using Zirconium beads on a high-

frequency oscillator (Precellys homogenizer, Bertig Instruments). The homogenates were

centrifuged at 6,000g for 10 min and the supernatant was collected for the extraction fatty acyl

lipid mediators using C18 solid phase extraction cartridges as described earlier [258-260]. Fatty

49

acyl lipid mediator extracts were subjected to LC-MS analysis essentially as described before

[258]. Under the LC-MS conditions employed, the detection limit for most of the lipid

mediators was 1 pg on the column and the quantitation limit was 5 pg on the column with a

signal/noise ratio >3. Tissue weights from each sample (range: 14-70 mg, average: 43 mg,

inter-quartile range: 32-56 mg) were used for normalization of the LC-MS data and the data are

reported as ng per gram of tissue.

3.3.7 Statistics:

Statistical analysis was performed using IBM SPSS Statistics Version 22 (IBM United Kingdom

Limited, Hampshire, England). Data were analyzed using a one-way repeated measures

ANOVA. The sphericity adjustment was checked, and if required, a Greenhouse-Geisser epsilon

correction was applied. In the instance, a metabolite verged achieving a main effect for time (P

< 0.1), a post-hoc analysis was run using a Bonferonni correction. Data are presented as mean

± standard error of the mean (SEM). Statistical significance was set at P < 0.05. This research

was an exploratory study and as such statistical power calculations based off a predicted mean

and standard deviation scores were unable to be completed. Sample size was determined by

the availability of existing muscle sample from a previous clinical trial exploring the effect of an

acute resistance exercise protocol on markers of post-exercise muscle damage [257].

50

3.4 Results:

3.4.1 Lipidomic analysis:

Lipid mediator profiles of human skeletal muscle tissue were generated via LC-MS. A total 129

unique lipid mediators were reliably detected (Supplemental Table 1, displayed in Appendix D).

However, many of these lipids were very lowly expressed or below the limits of quantification

of the assay (signal/noise ratio >3) in particular sample sets. Therefore, only lipid species that

were quantified in all post-exercise samples were statistically analysed. A main effect for time

was observed in a range of lipid-derived inflammatory mediators and their downstream

metabolites during post-exercise recovery. Peak concentration was generally identified at 2 h

post-exercise, however we were often unable to detect the specific time points at which these

changes became significant.

3.4.1.1 Cyclooxygenase pathway:

COX enzymes catalyse the first step in the conversion of AA to prostaglandins (PGE2, PGF2ɑ,

PGD2 and PGI2) and thromboxane (TXA2). TXA2 is a highly unstable and non-enzymatically

decomposes to TXB2 and 12(S)-HHTrE. Both metabolites serve as surrogate marker of TXA2

biosynthesis. Both of these inactive metabolites were detected within skeletal muscle tissue

and were significantly increased post-resistance exercise (P < 0.05) with the greatest

concentrations identified at 2 h post exercise (Figure 3.1). There were no significant changes

detected in any of the primary prostaglandins derived from AA. PGE2 and PGF2ɑ were the most

abundant prostaglandins in skeletal muscle tissue, detected at concentrations of 1.13 ng/g and

0.68 ng/g respectively at rest. Post-exercise, both PGE2 and PGF2ɑ were elevated 2 h post

exercise, however, the increase did not reach statistical significance (PGE2: P = 0.075, PGF2ɑ: P =

0.078) (Figure 3.1). PGD2 levels were very low and often below detectable limits, while PGI2

(measured as the stable non-enzymatic hydrolysis product 6-keto-PGF1α) was not detected.

Within muscle tissue, PGF2α and PGE2 were present at considerably higher concentrations

compared to their inactive circulating secondary (15-keto-PGE2/PGF2α) and tertiary (13,14-

dihydro-15-keto-PGE2/PGF2α) enzymatic degradation products. However, downstream

metabolites of other prostaglandin species were identified in muscle. A bioactive downstream

metabolite of the EPA-derived PGD3, 15-Deoxy-Δ12,14-prostaglandin J3, was detected at

concentrations of 2.93 ng/g in resting muscle and increased significantly during the acute

51

stages of post-exercise recovery with a peak response observed at 2 h post (P < 0.05) (Figure

3.1).

Figure 3.1: Metabolites of the cyclooxygenase pathway derived from arachidonic acid. Values depicted

are mean values ± SEM. *denotes a main effect for the exercise intervention (p < 0.05).

52

3.4.1.2 Lipoxygenase pathway:

Lipoxygenase isoform expression has been characterized in various cell types involved in acute

inflammation including neutrophils (5-LOX), platelets (12-LOX) and monocytes (15-LOX) [261].

5-LOX metabolises AA to 5-hydroperoxyeicosatetranoic acid (5-HpETE), which is either reduced

to 5-hydroxyeicosatetraenoic acid (5-HETE) or further metabolised to the potent neutrophil

chemoattractant leukotriene B4 (LTB4). In resting skeletal muscle tissue, LTB4 was below the

level of detection in the majority of samples analysed and when detected it was generally

present only at very low concentrations (≤1ng/g). However, LTB4 was consistently detected at

much more abundant levels within muscle tissue (~3ng/g) during post-exercise recovery,

specifically at the 2 h time point, however this failed to reach statistical significance (Figure

3.2). 5-HETE also verged on a time-dependent increase (P = 0.085). Post-hoc analysis verged on

a significant increase in intramuscular 5-HETE to concentrations of 8.99 ng/g at 2 h post-

exercise when compared to resting levels of 3.38 ng/g) (P = 0.058) (Figure 3.2). Further, 20-

COOH-LTB4, a downstream metabolite of LTB4 showed a significant increase, with the highest

response occurring at 2 h post exercise (P < 0.05) (Figure 3.2). In addition to AA, the 5-LOX

enzyme also has the capacity to oxidize the omega-3 fatty acids DHA and EPA.

Monohydroxylated-DHA (HDoHE) products of the 5-LOX pathway including 4-

hydroxydocosahexanoic acid (4-HDoHE) and 7-hydroxydocosahexanoic acid (7-HDoHE) were

detected in resting muscle at concentrations of 2.23 ng/g and 0.98 ng/g respectively. Both 4-

HDoHE and 7-HDoHE showed a significant increase with peak muscle concentrations of 3.76

ng/g and 1.54 ng/g respectively observed at 2 h post-exercise (P < 0.05) (Figure 3.3). The 5-LOX

product of EPA, 5-HEPE was similarly detected within skeletal muscle tissue but at

concentrations of 0.52 ng/g, approximately 10 times lower than the corresponding AA product

5-HETE. Intramuscular 5-HEPE was not significantly influenced by resistance exercise.

53

Figure 3.2: Metabolites of the lipoxygenase pathway derived from arachidonic acid. Values depicted are

mean values ± SEM. *denotes a main effect for the exercise intervention (p < 0.05). ^ denotes statistical

significance compared to pre-exercise values (p < 0.05).

54

Figure 3.3: Metabolites of the lipoxygenase pathway derived from docosahexaenoic acid. Values

depicted are mean values ± SEM. *denotes a main effect for the exercise intervention (p < 0.05).

The 12-LOX enzyme is expressed primarily in platelets and metabolizes AA to form 12-

hydroxyeicosatetraenoic acid (12-HETE), which can stimulate leucocyte chemotaxis and

platelet aggregation [262-264]. Additionally, in one key pathway of LX biosynthesis, 5-LOX

derived LTA4 is converted to LXA4 and LXB4 via the sequential action of platelet 12-LOX. 12-

HETE was by far the most abundant monohydroxylated-FA product detected in resting muscle,

present at concentrations of ~20 ng/g. We found 12-HETE increased with time (P < 0.05) with

peak concentrations of 63.81 ng/g observed at 2 h post-exercise (Figure 3.2). Similarly, muscle

tetranor 12-HETE, a downstream metabolite of 12-HETE increased significantly post-exercise (P

< 0.05), with the greatest response at 2 h post-exercise (Figure 3.2). The 15-LOX enzyme is

highly expressed in eosinophils, epithelial cells and alternatively activated M2 macrophages

and acts to convert AA to 15-hydroxyeicosatetranoic acid (15-HETE) [265]. While 15-HETE did

not achieve a main effect for time overall, post-hoc analysis showed a significant increase at 2

h post-exercise (6.50 ng/g) when compared to pre-exercise levels (3.84 ng/g) (P = 0.013)

(Figure 3.2).

LOX metabolites of the n-3 PUFA DHA have been established to be key pathway markers and

intermediates in the biosynthesis of SPMs. The 12-LOX enzyme converts the 22-carbon DHA to

14-hydroperxydocosahexanoic acid (14-HpDoHE) which is reduced to 14-HDoHE or

55

metabolised to MaR1/2 via further action of 12-LOX. Thus, 14-HDoHE is considered a key MaR

pathway activation marker. Similarly, 15-LOX converts DHA substrate to 17-H(p)DoHE, a key

intermediary step in the biosynthesis of both the D-series resolvins and protectins. We

detected 14-HDoHE in resting muscle at concentrations of 0.68 ng/g whereas 17-HDoHE was

not found to be present at detectable levels. There was no significant effect of exercise on 14-

HDoHE or 17-HDoHE. Nevertheless, peak intramuscular concentrations of 1.60 ng/g 14-HDoHE

were observed at the 2 h post-exercise time-point. MRM transitions corresponding to mature

SPMs including lipoxins (LXA4 and LXB4), E-series resolvins (RvE1 & RvE3), D-series resolvins

(RvD1, RvD2 and RvD5), protectins (PD1 and 10S,17S-DiHDoHE) and maresins (MaR1) were

additionally monitored by our LC-MS/MS assay. However, mature SPMs were not found to be

present at detectable levels locally within muscle tissue biopsies at rest or throughout 24 h

post-exercise recovery.

3.4.1.3 Epoxygenase pathway:

The cytochrome P-450 (CYP) enzymes metabolize n-6 PUFA AA to a family of

epoxyeicosatrienoic acid (EpETrE) regioisomers. Once formed, these EpETrEs are rapidly

metabolized by the soluble epoxide hydrolase (sEH) enzyme to form corresponding vicinal

diols, dihydroxyeicosatrienoic acids (DiHETrEs). 5,6-EpETrE was the only primary enzymatic

epoxy AA-metabolite that showed an effect for the exercise intervention, with concentrations

increasing 3-fold to ~15ng/g at 2h post-exercise (P < 0.05) (Figure 3.4). Whilst 8,9-EpETrE,

11,12-EpETrE, 14,15-EpETrE were also found in relative abundance in skeletal muscle tissue at

rest (~3-10 ng/g), they did not display a significant effect of exercise. Alternatively,

downstream products of the sEH enzyme, 11,12-DiHETrE and 14,15-DiHETrE were present at

lower absolute concentrations in resting muscle (~1 ng/g), but significantly increased in

response to exercise with a peak response at 2 h of recovery (P < 0.05) (Figure 3.4). 5,6-

DiHETrE and 8,9-DiHETrE regioisomers were also detected at low levels in resting muscle,

however no effect of the exercise intervention was observed.

CYP epoxidase enzymes also have the capacity to metabolise the n-6 linoleic acid to bioactive

products. Linoleic acid derived 9,10-DiHoME, is a leukotoxin generated by neutrophils during

56

the oxidative burst, and showed a main effect for the exercise intervention (Figure 3.5).

Similarly, 12(13)-DiHOME, an isoleukotoxin with wide-ranging effects in inflammation, showed

a main effect for the exercise intervention (Figure 3.5).

Figure 3.4: Metabolites of the epoxygenase pathway derived from arachidonic acid. Values depicted are

mean values ± SEM. *denotes a main effect for the exercise intervention (p < 0.05).

57

Figure 3.5: Metabolites of the epoxygenase pathway derived from linoleic acid. Values depicted are

mean values ± SEM. *denotes a main effect for the exercise intervention (p < 0.05).

3.5 Discussion:

The present study explored the intramuscular lipid mediator response following an acute bout

of resistance exercise. A total of 129 unique lipid mediators were identified within skeletal

muscle tissue. The post-exercise response was characterized by changes in lipid derivatives of

the COX-1 and -2, LOX (5-, 12-, 15-LOX), and CYP pathways. Peak induction of both pro-

inflammation and SPMs occurred at 2 h post-exercise. The findings from this study identify a

complex lipid response to resistance exercise that may play an essential role in regulating the

onset and resolution of acute exercise-induced skeletal muscle inflammation.

This was the first paper to use a targeted lipidomics approach to extensively characterize the

lipid mediator profile of skeletal muscle tissue at rest and in response to acute exercise. The

majority of human research exploring the effect of exercise on lipid species has focused

primarily on a select few prostaglandins, specifically PGE2 and PGF2ɑ, likely due to their

complex role in regulating acute inflammation, perceptions of pain and purported role in

muscle cell growth/regeneration [132-135]. Although some studies have reported elevated

circulating levels of PGE2 following exercise-induced muscle injury [102, 117-119], these

findings have not yet been replicated in skeletal muscle tissue [93, 132]. PGF2ɑ has been shown

58

to increase locally within skeletal muscle tissue following both eccentric [132] and isotonic

resistance exercise protocols [133] and plays a significant role in in-vitro myofibre hypertrophy

[266] and post-exercise muscle protein synthesis [107]. Further, recent research from our

group found a significant increase in serum levels of the circulating PGF2ɑ metabolite 15-keto-

PGF2ɑ early (1 h) following resistance exercise in humans [102]. While the increase in muscle

PGE2 or PGF2ɑ failed to reach statistical significance, they were the most abundant AA derived

PGs detected within muscle and both markers showed a strong trend for an effect of the

exercise intervention (P = 0.075 and P = 0.078 respectively). Further, increase in TXA2

(measured as TXB2 and 12(S)-HHTrE) post-exercise with peak responses at the 2 h time-point

may reflect a pro-aggregatory, pro-thrombotic circulatory effect that occurs in response to

both maximal aerobic [267] and resistance exercise protocols [102].

The other major metabolic pathway leading to the formation of lipid species involved in the

regulation of inflammation is the lipoxygenase pathway. Increases in the 5-LOX derived LTB4

has been reported in blood serum samples following acute resistance exercise [102] and high

speed running [268]. LTB4 is a potent neutrophil chemoattractant and a powerful stimulator of

vasoconstriction and blood vessel permeability [269]. Expression of 5-LOX is essentially limited

to bone-marrow derived cells including inflammatory neutrophils and

monocytes/macrophages [270]. It is therefore not surprising that in the present study LTB4 was

very lowly expressed in skeletal muscle tissue prior to exercise. At 2 h post-exercise, LTB4 was

detectable in the majority of subjects, and a downstream derivative, 20-COOH LTB4,

demonstrated a main effect for the exercise intervention. A less well-described branch of the

5-LOX pathway involves the metabolism of n-3 PUFA DHA to form mono-hydroxylated fatty

acids 4-HDoHE and 7-HDoHE. These fatty acids were found in abundance in skeletal muscle

tissue and increased during post-exercise recovery, however no such change was observed

previously in blood serum samples [102].

In addition to 5-LOX, metabolites of the human platelet type 12-LOX enzyme 12-HETE and its

downstream derivate tetranor 12-HETE were also elevated within muscle during post-exercise

recovery. Both metabolites are pro-inflammatory in nature and have been shown to act

59

transcellularly to modify the responsiveness of neutrophils to other chemotactic factors [271].

We previously observed a similar increase in 12-HETE and downstream tetranor 12-HETE in

human blood serum samples during recovery from resistance exercise [102]. Interestingly 12-

LOX expressing platelets have been implicated in the transcellular biosynthesis of pro-

resolution LX mediators through interactions with 5-LOX expressing PMNs. This pathway

involves leucocyte-platelet interactions during which the 5-LOX derived leukotriene

intermediate LTA4 is taken up by 12-LOX expressing platelets for subsequent conversion to

LXA4 [272, 273]. Further, the 15-LOX pathway has been implicated as a second the endogenous

route of LX biosynthesis species. 15-LOX is highly expressed in alternatively activated

macrophages and epithelial cells. The secretion of 15-LOX product 15-HETE can be taken up by

5-LOX expressing cells and converted to LXA4 and LXB4 [274]. In this study we demonstrated a

significant increase in 15-HETE at 2 h post-exercise, a finding which has been replicated in

human blood serum samples [102]. Our finding of simultaneous induction the primary

products of both the 5-LOX/12-LOX and 15-LOX/5-LOX pathways locally within skeletal muscle

during post-exercise recovery would presumably be a permissive environment for local LX

biosynthesis to occur. However, LXA4 or LXB4 were unable to be detected locally within muscle

biopsy homogenates. Nevertheless, these local changes within the exercised musculature

suggests that skeletal muscle tissue may potentially contribute to the exercise-induced

systemic lipoxin response previously reported [102].

The CYP pathway is a third, perhaps less well-known, branch of the AA metabolic pathway. The

increase in epoxyeicosatrienoic acid regioisomer 5-6-EpETrE and dihydroxyeicosatrienoic acids

11,12-DiHETrE and 14,15-DiHETrE in skeletal muscle supports observations made in serum

samples when measured during the early stages of post-exercise inflammation [102]. The

physiological function of these derivatives remains unexplored in skeletal muscle tissue.

However, in vascular smooth muscle and endothelium cells they play anti-inflammatory roles

through in the inhibition of prostaglandin and cytokine induced inflammatory responses [275].

CYP enzymes also metabolise linoleic acid via epoxidation to form epoxyoctadecmonoeic acids

(EpOMEs). EpOMEs are rapidly hydrolysed by the sEH enzyme to form DiHOMEs [276]. EpOMEs

and DiHOMEs are leukotoxins that play a role in the suppression of neutrophil respiratory burst

activity, vasodilation and cellular apoptosis [277, 278]. A cycling based intervention comprising

60

of a 75km time trial had no effect on 9-10,DiHOME 1.5 h and 21 h post-exercise in plasma

samples of competitive road cyclists [277]. Alternatively, a bout of acute resistance exercise

triggered an increase in 9(10)-EpOME and 9-10,DiHOME in serum samples [102]. Results from

the present study showed an increase in 9-10,DiHOME and 12(13)-DiHOME suggesting that

discrepancies in the previous literature may be due to the differences in the type of exercise

performed and the training status of the subjects used in each study.

Collectively, these findings demonstrate an increase in lipid derived mediators of the COX, LOX

and CYP pathways during post-exercise muscle recovery. The concept of a biologically active

inflammatory resolution programme governed by lipid derivatives was first proposed in a TNF-

ɑ-stimulated model of acute inflammation in the murine air pouch [185]. Within this model,

early formation of LTB4 and PGE2 at the onset of inflammation was succeeded by a class-

switching of eicosanoids to LXA4. During this process, interactions between inflammatory and

host tissue cells enabled the biosynthesis of resolution mediators [185]. Alternatively, in a

model of zymosan-A stimulated murine peritonitis, the onset of inflammation was

characterized by a concomitant increase in LTB4 and LXA4 followed by a late appearance of

PGE2 at the onset of resolution [186]. These findings demonstrate that the temporal regulation

of lipids and their role in inflammation is likely cell-type and stimulus specific. Recent work

from our group profiled the human lipid response to acute resistance exercise in serum

samples [102]. This study showed an increase in key prostaglandin, leukotriene, lipoxin and

resolvin species during the early stages of acute inflammation (1-3 h), followed by an increase

in 15-LOX derivatives and some prostaglandin metabolites (6-keto-PGF1ɑ and 13,14dh-

15kPGE2) at 24 h post- exercise [102]. One of the limitations of the present study was the lack

of statistical power preventing post-hoc analysis of the observed effect of the exercise

intervention. However, for all intramuscular lipid derivatives peak expression occurred at 2 h

post-exercise, which appears consistent with findings from serum samples [102]. Interestingly

lipid species that require transcellular interactions, including lipoxins, resolvins and protectins,

were either undetected, or very lowly expressed in skeletal muscle tissue. These lipids were

detectable previously in abundance in human serum samples during post exercise recovery

and are known to play a vital role in the active resolution of acute inflammation [102].

61

3.6 Conclusion:

This is the first study to profile the lipid mediator response to acute resistance exercise in

skeletal muscle tissue. An increase in lipids derived from the COX, LOX and CYP pathways was

observed during the early stages of post-exercise muscle recovery. Peak induction of AA

derived classical pro-inflammatory markers PGE2, PGF2ɑ, and both upstream and downstream

markers of LTB4 occurred at 2 h post-exercise. Further various derivatives of the 5-LOX (4-

HDoHE and 7-HDoHE), 12-LOX (12-HETE and tetranor 12-HETE) and 15-LOX (15-HETE)

pathways were identified in abundance in skeletal muscle tissue post-exercise and may

resemble transient cellular intermediates for the formation of pro-resolution lipoxin and

resolvin species. In alternative models of acute inflammation, these lipids are involved in

coordinating a biologically active inflammatory resolution programme that has been

mechanistically linked to processes tissue healing. Future research exploring the physiological

significance and function of these intramuscular lipids in both skeletal muscle and

serum/plasma samples post-exercise, will be useful in further characterizing the significance of

the intramuscular inflammatory response in post-exercise muscle recovery.

3.7 Acknowledgments:

The authors would like to acknowledge to assistance of Associate Professor John Reynolds for

his contributions in the statistical analysis of the dataset. We further thank Dr. Andrew

Garnham (Deakin University) for all muscle biopsy procedures, and the participants for

volunteering their time to be involved in the trial.

3.8 Disclosures:

This research was conducted in the absence of any commercial or financial relationships that

could be construed as a potential conflict of interest.

62

63

CHAPTER 4:

Ibuprofen supplementation and its effects on NF-κB activation in skeletal muscle

following resistance exercise.

Vella L, Markworth JF, Peake JM, Snow RJ, Cameron-Smith D, Russell AP. Ibuprofen

supplementation and its effects on NF-κB activation in skeletal muscle following resistance

exercise. Physiological Reports, 2014 Oct 24; 2:10. doi: 10.14814/phy2.12172 (Appendix B).

64

4.1 Abstract:

Purpose: Resistance exercise triggers a subclinical inflammatory response that plays a pivotal

role in skeletal muscle regeneration. Nuclear factor-κB (NF-κB) is a key stress signaling

transcription factor that regulates acute and chronic states of inflammation. The classical NF-

κB pathway regulates the early activation of post-exercise inflammation; however there

remains scope for this complex transcription factor to play a more detailed role in the muscle

regeneration process. Methods: Sixteen volunteers completed a single bout of lower body

resistance exercise with the ingestion of three 400 mg doses of ibuprofen or a placebo control.

Muscle biopsy samples were obtained prior to exercise and at 0, 3 and 24 h post-exercise.

Samples were analysed for key markers of NF-κB activity. Results: Phosphorylated p65 protein

expression and p65 inflammatory target genes were elevated immediately post-exercise.

These responses were independent of the two treatments. These changes did not translate to

an increase in p65 DNA binding activity. NF-κB p50 protein expression and NF-κB p50 binding

activity were lower than pre-exercise at 0 and 3 h post-exercise, but were elevated at 24 h

post-exercise. Conclusion: These findings provide novel evidence that two distinct NF-κB

pathways are active in skeletal muscle after resistance exercise. The initial wave of activity

involving p65 resembles the classical NF-κB pathway and is associated with the onset of an

acute inflammatory response. The second wave of NF-κB activity comprises the p50 subunit,

which has been previously shown to resolve acute inflammation in rodent inflammatory

models. The current study showed no effect of the ibuprofen treatment on markers of the NF-

κB pathway, however examination of the within group effects of the exercise protocol suggests

that this pathway warrants further research.

65

4.2 Introduction:

Unaccustomed resistance exercise causes skeletal muscle damage that impairs muscle function

and promotes sensations of pain. The precise signaling mechanisms that initiate muscle repair

following exercise-induced damage remain a topic of ongoing debate (previously reviewed by

Paulsen et al. [279]). The onset of muscle damage triggers a complex interplay of intracellular

events that involve myofibre injury, acute inflammation and cellular repair. At a symptomatic

level, inflammation in skeletal muscle is characterised by swelling and soreness [279].

Consequently post-exercise inflammation is often ascribed as a cause of delayed-onset muscle

soreness (DOMS). DOMS typically occurs 2448 hours post-exercise, and is accompanied by a

secondary reduction in muscle force-generating capacity. Treatment of DOMS has focused on

reducing inflammation; however this may be detrimental to processes of cellular repair [103].

Attenuating exercise-induced inflammation using non-steroidal anti-inflammatory drugs

reduces skeletal muscle protein synthesis [280] and impairs satellite cell proliferation [87]. To

develop more efficacious treatments for DOMS, a better understanding of the mechanisms

that govern inflammation and tissue repair in skeletal muscle after exercise is required.

The nuclear factor-kappa B (NF-κB) transcription factor acts as a central regulator of

inflammatory signaling pathways. The NF-B family consists of five subunits, including NF-κB1

(p105/p50), NF-κB2 (p100/p52), RelA (p65), RelB and cREL that share an amino terminus Rel

homology domain. The Rel domain permits DNA binding, nuclear localization, dimerization and

interaction with its own inhibitory protein IκB [190, 191, 194]. Under basal conditions RelA

(p65), cREL and RelB subunits remain sequestered within the cytoplasm bound to an inhibitory

IB protein. The IB family that regulates NF-B includes the subunits IκBβ, IκBα, IκBγ IκBε, and

Bcl-3. Unlike the Rel proteins, subunits p50 and p52 are synthesized as large precursor proteins

(p105 and 100, respectively) that require proteolytic processing to permit nuclear localization.

These subunits lack a REL domain, to initiate gene transcription, and hence are primarily

considered as repressors of gene transcription [194, 281].

66

Upon stimulation, the IκB kinase complex (IKK) controls the degradation of IκB and its

precursor proteins, thereby enabling the NF-κB REL subunits to control gene transcription. The

IKK complex consists of two catalytic kinases IKKα and IKKβ, and a regulatory IKKγ subunit.

IKKβ activates the classical NF-κB signaling pathway through the phosphorylation and

subsequent degradation of the inhibitor IκBα [192, 199]. The classical pathway typically

comprises p65:p50 heterodimers, and is essential for the activation of acute inflammation by

controlling the transcription on inflammatory cytokines and acute phase proteins [192, 199].

More recently an alternative NF-κB pathway has been described, which is dependent on IKKα

[282]. Alternative signaling involves the activation of a secondary NF-κB inducing kinase (NIK),

and has been linked to the activation of both p52 and p50 subunits. The functional significance

of IKKα-dependent gene expression in acute inflammation is not yet well established.

However, preliminary research in rat muscle tissue suggests p50 homodimers may play a

crucial role in the resolution of acute inflammation [283, 284]. Transgenic rodents expressing

enhanced p50-p50 DNA binding showed reduced expression of inflammatory cytokines and

impaired leucocyte recruitment during inflammatory resolution [285]. Further, p50-p50

complexes bind to distinct κB response elements in COX-2 promoter regions, and have shown

to regulate COX-2 expression through association with transcriptional co-activators of the

C/EBP family [286, 287]. These findings implicate that a biphasic NF-κB activation patterns may

represent a key regulatory pathway in the expression of COX derived inflammatory resolution

mediators, as identified in chapter 3.

Despite the importance of inflammation in tissue regeneration post-exercise, very little is

known about how NF-κB activity is regulated in skeletal muscle after acute exercise. A transient

increase in various components of the classical NF-κB signaling pathway is observed in rat

muscle post-exercise [233, 234, 288-290]. In contrast, a decrease in NF-κB DNA binding at 0

hours post-exercise with a return to near baseline 1 hour post-exercise was observed in human

muscle following a traditional resistance exercise model [238]. Recent work from our

laboratory, using a similar resistance exercise model, demonstrated an increase in NF-κB

binding to the promoter region of key inflammatory cytokines at 2 hours post-exercise, with a

67

return to baseline levels at 4 hours post-exercise [239]. These findings suggest that a transient

NF-κB response contributes to acute post-exercise inflammation.

To enhance our understanding of the cellular mechanisms that regulate both the onset and

resolution of post-exercise inflammation, the current study aimed to investigate changes in the

activity of the subunits that comprise the classical and alternative NF-κB signaling pathways. It

was hypothesized that the regulation of NF-κB post-exercise would involve two distinct waves

of activation. Specifically, we hypothesized that the classical NF-κB pathway (involving p65-p50

heterodimers) would be activated soon after exercise during the early phases of inflammation,

while the alternative pathway (comprising p50 homodimers) would be activated at later time

points after exercise that correspond with the resolution of acute inflammation [284, 285]. It

was further hypothesized that the administration of ibuprofen would blunt both waves of NF-

κB activation, providing a potential mechanism through which anti-inflammatory medication

might attenuate exercise-induced inflammation in skeletal muscle.

68

4.3 Methods:

4.3.1 Participants:

As previously described in chapter 2, sixteen healthy male subjects were recruited to

participate in the study (characteristics shown in Table 1) [102]. All participants completed a

medical history questionnaire that was used to identify and exclude participants with a

diagnosed condition or illness that prevented them from completing strenuous exercise.

Exclusion criteria included participation in a lower body resistance exercise program within the

last 6 months to ensure a muscle damage response from the exercise stimulus, and/or chronic

treatment with anti-inflammatory drugs. Current use of prescription medication or nutritional

supplements also excluded subjects from participating.

Table 4.1: Subject characteristics and strength testing data. Values are mean values ± SEM. No

significant differences were observed between the two groups.

Characteristics Strength (1RM)

Age (y) Height (m) Body mass

(kg) BMI Squat (kg) Leg Press (kg)

Leg

Extension

(kg)

PLA 23.9

± 1.3

1.89

± 0.1

86.9

± 4.5

24.5

± 1.2

94.9

± 5.4

237

± 17

236

± 18

IBU 23.0

± 0.5

1.89

± 0.1

89.1

± 4.4

24.8

± 0.8

91.9

± 6.0

240

± 15

196

± 22

4.3.2 Ethics approval:

Prior to participation each subject received written and oral information regarding the nature

of the experiment before providing written consent to participate. All procedures involved in

this study were approved by the Deakin University Human Research Ethics Committee

(DUHREC 2010-019). All muscle sampling procedures were performed in accordance with the

Helsinki declaration.

69

4.3.3 Familiarization and strength testing:

Each participant completed a familiarization session at least 7 days prior to completing the

exercise trial. Participants performed repetition maximum testing to determine the

experimental exercise load (80% of 1 repetition maximum (1RM)). The maximal weight each

subject could lift was determined for the Smith machine-assisted squat, the leg press, and the

leg extension. These data were substituted into the validated Brzycki equation to predict 1RM

for each participant [244, 291]. The participants were required to abstain from any further

exercise until completion of the trial.

4.3.4 Experimental procedures:

The participants reported to the laboratory in an over-night fasted state, having abstained

from caffeine, tobacco and alcohol for the preceding 24 h. Following 30 min of supine resting a

pre-exercise muscle biopsy was taken. Participants then completed a 10 min warm up

consisting of low intensity cycling on a bicycle ergometer, and one low resistance set for each

exercise at approximately 30-50% of the participants 1RM. Each participant then completed a

single bout of intense resistance exercise. This session consisted of 3 sets of 8-10 repetitions of

a bilateral Smith machine assisted squat, 45-degree leg press and leg extension. These

exercises were all performed at 80% 1RM. The exercises were performed sequentially as a

circuit, with 1 min rest between each exercise, and 3 min rest between sets. We have

previously used this exercise protocol, and demonstrated that it activates inflammatory

signaling pathways [239]. Following the completion of the exercise bout, the participants

rested in a supine position while muscle biopsy samples were collected. The participants

returned to the laboratory the following morning in an over-night fasted state for a final

muscle biopsy sample. Standardized meals were provided to participants the night preceding

the trial (carbohydrate 57%, fat 22%, protein 21%), in the laboratory immediately following the

3 h muscle biopsy (carbohydrate 71%, fat 13%, protein 16%), and an evening meal

(carbohydrate 64%, fat 27%, protein 18%).

70

4.3.5 NSAID administration:

Prior to the exercise bout, the participants were randomly assigned to either the ibuprofen

(NSAID) group (n = 8) or the placebo group (PLA) (n = 8). Participants in the NSAID group

consumed the maximum recommended over-the-counter dose of 1200 mg of ibuprofen as

three doses of 400 mg throughout the trial day. The first dose was administered upon arriving

to the laboratory on the first morning of the trial, immediately prior to the first muscle biopsy

sample. Participants were instructed to consume two additional doses at 2:00pm and 8:00 pm

the same evening. This dosing structure was prescribed to optimise levels of circulating

ibuprofen to biologically active levels throughout the course of the trial day. This protocol has

previously been validated by our research group in this same group of study participants [102].

Alternatively, the placebo consumed a gelatin capsule containing powdered sugar, identical in

appearance to the ibuprofen capsule.

4.3.6 Sample collection:

Muscle biopsy samples were obtained under local anaesthesia (Xylocaine 1%) from the vastus

lateralis muscle of either leg using the percutaneous needle biopsy technique modified to

include suction [245]. Samples were obtained prior to exercise, immediately following exercise

and at 3 h and 24 h post-exercise. The muscle biopsy sample obtained immediately post-

exercise was taken within 1-2 min of the completion of the exercise protocol, and will herein

after being referred to as 0 h post-exercise. The muscle biopsy procedure has been shown to

trigger a local inflammatory response [62]. To minimize interference from the biopsy

procedure, samples prior to exercise and at 24 h post-exercise were taken from the same leg,

while muscle samples obtained at 0 and 3 h post-exercise were taken from the opposite leg.

Samples within the same leg were taken at least 5 cm from the previous site. We have

previously reported that this technique is effective for minimizing cytokine gene expression

and NF-B activity in response to muscle biopsies [239]. Excised tissue was immediately

immersed in liquid nitrogen and stored at 80°C until further analysis.

71

4.3.7 Protein extraction and quantification:

Muscle samples were homogenized in ice cold RIPA buffer (50 mM Tris-HCl, pH 7.4, 150 mM

NaCl, 0.25% deoxycholic acid, 1% NP-40, 1 mM EDTA supplemented with protease and

phosphatase inhibitors including 1 mM PMSF, 1 µg/ml aprotinin, 1 µg/ml leupeptin, 1 mM

Na3VO4 & 1mM NaF). The homogenate was agitated for 1 h at 4°C and centrifuged at 13,000

g at 4°C for 15 min. The resultant supernatant was removed and stored at 80°C until further

analysis. Total protein concentration was determined using a BCA protein assay kit according

to the manufacturer’s instruction (Pierce, Rockford, IL). Protein samples (50 µg) were

denatured in loading buffer and separated by a 10% SDS-PAGE and transferred to a PVDF

membrane. Membranes were blocked for 90 min at room temperature in 5% BSA/Tris buffered

saline with 0.1% Tween 20 (TBST). Primary antibodies [phosphorylated NF-κB p65 Ser536, total

NF-κB p65, NF-κB p100/p52, NF-κB p105/p50, NF-κB cREL and β-actin (all obtained from Cell

Signaling Technologies, Arundel, QLD)] were diluted to 1:1,000 in 5% BSA/TBST, applied and

incubated overnight at 4°C with gentle agitation. Membranes were washed for 30 min in TBST

and probed with HRP conjugated secondary antibodies diluted to 1:2,000 in 5%BSA/TBST, and

incubated for 1 h at room temperature. Proteins were visualised by using Western Lighting

enhanced chemiluminescence (Perkin Elmer Lifesciences, Boston, MA). Signals were captured

using a Kodak Digital Image Station 2000M (model: 440CF; Eastman Kodak, Rochester, NY) and

quantified by densitometry band analysis using Kodak Molecular Imaging Software (Version

4.0.5, © 1994-2005, Eastman Kodak). Phosphorylated NF-κB p65 protein was normalized to

total p65 protein (Supplementary Figure 4.1A). NF-κB p50 protein expression was normalized

to its precursor protein p105 (Supplementary Figure 4.1B). NF-κB p52 and cREL protein was

normalized to β-actin (Supplementary Figure 4.1C).

4.3.8 RNA extraction and RT-PCR:

Total cellular RNA was extracted as previously described [257] using the ToTALLY RNA Kit

(Ambion, Austin, TX). RNA quality and concentration were determined using the Agilent 2100

Biolanalyzer (Agilent Technologies, Palo Alto, CA). First strand cDNA was generated from 0.5 µg

total RNA using the AMV RT kit (Promega, Madison, WI). RT-PCR was performed in duplicate

using the Biorad CFX384 system (Biorad, Hercules, CA), containing 5XHOT FirePol® EvaGreen

Mix (Integrated Science, Sydney, NSW), forward primer, reverse primer, sterile nuclease free

72

water and cDNA (0.125 ng/μL). Data were analysed using a comparative critical threshold (Ct)

method, where the amount of the specified target gene normalised to the amount of

endogenous control, relative to the control value is given by 2-ΔΔCt. The endogenous control

used in this experiment was GAPDH. The efficacy of GAPDH as an endogenous control was

examined using the equation 2-ΔCt. Primers were designed using Primer Express software

package version 3.0 (Applied Biosystems). Gene sequences were obtained from GenBank

(Table 2). Primer sequence specificity was confirmed using BLAST. A melting point dissociation

curve was generated by the PCR instrument for all PCR products to confirm the presence of a

single amplified product.

Table 4.2: Primer sequences were designed using Primer Express Software v 3.0 (Applied

Biosystems) using sequences accessed through Genbank and checked for specificity using

nucleotide-nucleotide BLAST search.

Gene Accession No. Primer Sequence

GAPDH NM_21130 Forward CAT CCA TGA CAA CTT TGG TAT CGT

Reverse CAG TCT TCT GGG TGG CAG TGA

MCP-1 NM_002982 Forward TCC CAA AGA AGC TGT GAT CTT CA

Reverse CAG ATT CTT GGG TGG AGT GA

IL-6 NM_000600 Forward GCG AAA GGA TGA AAG TGA CCA T

Reverse AGA CAA GCC CAG CAA TGA AAA

IL-8 NM_000584 Forward CTG GCC GTC GCT CTC TGG

Reverse TTA GCA CTC CTT GGC AAA ACT

TNF-α X_02910 Forward GGA GAA GGG TGA CCG ACT CA

Reverse TGC CCA GAC TCG GCA AAG

COX-2 U_20548 Forward GAA TCA TTC ACC AGG CAA ATT G

Reverse TGG AAG CCT GTG ATA CTT TCT GTA CT

4.3.9 Nuclear extraction and Transcription Factor (TF) Assay:

Nuclear and cytoplasmic proteins were extracted from 20 mg of muscle tissue using a NE-PER

Nuclear and Cytoplasmic Extractions Reagents (Pierce, Rockford, IL), according to the

manufacturer’s instructions. Western blot analysis probed for GAPDH, α-tubulin and Lamin A

was performed to ensure the nuclear extract was not contaminated by cytoplasmic proteins

73

(Supplementary Figure 2). A Transcription Factor Assay detecting specific transcription factor

DNA binding activity was performed according to manufacturer’s instructions (Cayman

Chemical, Ann Arbour, MI). Briefly, 10 µg of nuclear protein were loaded into a well containing

an immobilized NF-B consensus sequence (5’GGGACTTTCC-3’) and incubated overnight at

4°C. Primary antibodies for NF-B subunits p65 and p50 were loaded in to each well and

incubated at room temperature for 1 h. Each well was flushed using a diluted wash buffer for

30 min. Following a secondary 30 min wash, samples were incubated with secondary HRP-

conjugated antibody for a further 1 h at room temperature. To quantify transcription factor

binding, a developing solution containing a 3,3’,5,5’-Tetramethhylbenzidine (TMB) solution was

added to each well and incubated for 45 min at room temperature. DNA binding was then

quantified using photospectrometry with absorbance measurements taken at 450 nm using a

Multiscan RC plate reader (Labsystems, Finland) and Gen5 Data Analysis Software (BioTek,

Winooski, VT).

4.3.10 Statistics:

Data are expressed as means ± SEM. Prior to analysis the data displaying a lack of normality

were log transformed to stabilise variance. Data were analysed using a two-way ANOVA with

repeated measures for time. The sphericity adjustment was checked, and if required, a

Greenhouse-Geisser epsilon to the residual degrees of freedom was applied. A data point was

considered to be a statistical outlier where a z-score exceeded a pre-determined threshold of ±

4.00. Where appropriate we explored within-group pair-wise comparisons between individual

time-points using the Least Significant Differences of means at P < 0.05 to determine

statistically significant changes [292, 293]. Statistical analyses were performed using GenStat

for Windows 16th Edition (VSN International, Hemel Hempstead, UK).

74

4.4 Results:

4.4.1 Protein expression:

Expression of NF-κB phosphorylated p65 (Ser 536) increased over time (time effect p=0.006),

but this response was overall not different between the groups (interaction effect p=0.253,

treatment effect p= 0.176). Nevertheless, LSD pairwise comparisons indicated that the main

increase from baseline was in the placebo group at 0 h post-exercise. Within the placebo group

phosphorylated p65 protein expression remained elevated at 3 h post-exercise and returned to

baseline by 24 h (Figure 4.1A). No significant changes were observed in the ibuprofen group by

LSD comparisons. There was no effect of time or treatment for NF-κB total p65 (Figure

Supplementary Figure 4.1A). There was a trend toward a significant time treatment

interaction (p=0.057) for the protein expression of the p50 subunit, although a main effect for

time (p=0.376) or treatment (p=0.865) was not achieved. Further analysis revealed a statistical

outlier in the ibuprofen group at the 24 h time point. When this subject was removed from the

analysis, this trend was weaker (p=0.115) (Figure 4.1B). LSD comparisons indicated an increase

in p50 protein expression was observed at 24h post exercise in the placebo group only (Figure

4.1B). There were no main or interaction effects for the NF-κB p50 precursor protein p105

(Supplementary Figure 4.1B) or for p52 and cREL subunits (Figure 4.1C and Figure 4.1D).

75

Figure 4.1: Protein expression of NF-κB subunits p65, p50, p52 and cREL. Representative Western blots

for p-p65 (A), p50/p105 (B), p52 (C) and cREL (D) measured in muscle biopsy samples. Data are mean

arbitrary units ± SEM. * denotes statistical significance from pre-exercise values in the placebo group; ^

denotes statistical significance from 24 h post-exercise in the placebo group (p<0.05). White bars =

placebo group; black bars =ibuprofen group.

76

Supplementary Figure 4.1: Protein expression of total NF-κB p65, NF-κB p105 and β-actin. Data

are mean arbitrary units ± SEM. White bars = placebo group; black bars =ibuprofen group.

4.4.2 Transcription Factor assay:

To determine subunit-specific DNA binding of NF-κB following exercise, transcription factor

binding assays were performed for p50 and p65 subunits. NF-κB p50 showed a main effect for

time (p=0.035), with no time treatment interaction (p=0.851) or main effect for treatment (p

=0.488) (Figure 4.2B). LSD comparisons identified a significant increase in p50 binding that

occurred at 24 h post-exercise when compared to 0 and 3 h post-exercise within the placebo

group. This coincided with the increase in p50 protein expression (Figure 4.1B). No change in

p65 DNA binding was observed, despite an increase in phosphorylated p65 protein expression

(Figure 4.2B).

77

Figure 4.2: NF-κB subunits p65 (A) and p50 (B) binding to nuclear protein. Data are mean arbitrary units

± SEM. # denotes statistical significance from 0 h post-exercise in the placebo group; $ denotes

statistical significance from 3 h post-exercise in the placebo group. White bars = placebo group; black

bars =ibuprofen group.

4.4.3 RT-PCR:

The mRNA levels of IL-6 (Figure 4.3A), IL-8 (Figure 4.3B) and MCP-1 (Figure 4.3C) demonstrated

a main effect for time (p<0.01), with the highest elevation in expression levels at 3 h post-

exercise after significant increases at 0 h post-exercise. TNF-α mRNA remained unchanged

after exercise (Figure 4.3D). COX-2 mRNA showed a main effect for time (p<0.01), increasing at

0, 3 and 24 h post-exercise (Figure 4.3E). There were no significant interaction effects or main

effects for treatment for the inflammatory cytokines or COX-2 mRNA expression. Consistently,

LSD pairwise comparisons revealed similar changes over time from baseline within both

groups, demonstrating no effect of the ibuprofen supplementation protocol on exercise-

induced cytokine expression.

78

Figure 4.3: RT-PCR analysis of NF-κB target genes IL-6 (A), IL-8 (B), MCP-1 (C), TNF-α (D) and COX-2 (E) in

skeletal muscle cDNA. Data are mean arbitrary units ± SEM. * denotes statistical significance from pre-

exercise in the same treatment group; # denotes statistical significance from 0 h post-exercise in the

same treatment group; ^ denotes statistical significance from 24 h post-exercise in the same treatment

group (p<0.05). White bars = placebo group; black bars =ibuprofen group.

79

4.5 Discussion:

The current study aimed to explore the regulation of the different NF-κB subunits following a

single bout of lower body resistance exercise, and investigate the mechanism through which

ibuprofen treatment may influence post-exercise inflammation. The results of this study

provide preliminary evidence that an alternative NF-κB signaling pathway comprising

predominately p50 subunits is activated 24 h post-exercise. This pathway appears distinctly

different to the activation of NF-κB p65 subunit that occurs during the early stages of acute

post-exercise inflammation.

Phosphorylated NF-κB p65 protein expression was significantly elevated in the placebo group

when measured at 0 and 3 h post-exercise, and returned to baseline levels at 24 h post-

exercise. These findings support those of previous research showing a post-exercise increase in

key components of the classical NF-κB signaling pathway in skeletal muscle, including

phosphorylated NF-κB p65 protein [234, 239], phosphorylated IκBα protein [233, 239] and NF-

κB p65 DNA binding activity [236, 294]. In the present study, the increase in phosphorylated

NF-κB p65 protein expression was not associated with an increase in p65 transcription factor

binding activity at 0, 3 and 24 h post-exercise. However, NF-κB regulated cytokine genes

including MCP-1, IL-6 and IL-8 were increased at 3 h post-exercise. Previous work from our

group showed a transient increase in NF-κB p65 binding to the promoter region of genes

coding for inflammatory cytokines at 2 h after traditional resistance exercise that returned to

basal levels at 4 h [239]. This research performed a series of electrophoretic mobility shift

assays that looked specifically NF-κB binding to genes coding MCP-1, IL-6 and IL-8 and thus

differences in the methodology, particularly the timing of muscle biopsy samples and the

different experimental methods used to quantify DNA binding, may explain conflicting results.

Research models using an eccentric resistance exercise model could demonstrate an increase

in p65 DNA binding to nuclear protein at 3 h post-exercise [294, 295]. By contrast, research

from Durham et al showed a decrease in NF-κB DNA binding activity immediately post-

exercise, which returned to baseline levels at 1 h post-exercise [238]. Collectively, these

findings suggest that the activation of NF-κB DNA binding occurs transiently within the first few

hours of exercise. Changes in NF-κB activity in skeletal muscle may depend on the mode of

exercise and training status of participants.

80

This research also provides novel evidence for a potential role of NF-κB p50 activation as a

component of a biologically active inflammation-resolution program as outlined in chapter 3.

In our initial analysis of p50 protein expression, there was a trend towards a time group

interaction effect (p=0.057) but this trend was weaker (p=0.115) after removing a statistical

outlier. Despite this weaker interaction, within-group analysis by LSD comparisons revealed

that p50 protein expression was elevated in the placebo group at 24 h post-exercise.

Coincident with this response, p50 transcription factor binding activity was highest at 24 h

post-exercise. The biological significance of this delayed increase in NF-B p50 signaling during

the latter stages of post-exercise recovery remains uncertain. However, in vitro work suggests

that it may play a role in regulating the active resolution of acute inflammation in skeletal

muscle [282, 283, 296]. In a model of carageenin-induced pleurisy in rats, Lawrence et al [283]

provided preliminary evidence for a complex interplay between distinct NF-κB signaling

pathways that control an acute and transient inflammatory response. They reported that the

preliminary phase of NF-κB activation that occurred at 6 h was characteristic of the classical

NF-κB pathway, and was associated with the onset of acute inflammation. The secondary

phase was typical of the alternative NF-κB pathway, comprising p50 homodimers. Importantly,

inhibiting this wave of activity caused a prolonged inflammatory response, delaying the

resolution of key inflammatory mediators [283]. Findings from the present study support the

concept of two functionally distinct waves of NF-κB activity following traditional resistance

exercise. A primary wave consistent with classical NF-κB signaling pathways was observed

during the early stages of post-exercise muscle recovery and the onset of inflammation. A

secondary rise in NF-κB p50 expression occurred at 24 h post-exercise, at a time-point that

could be indicative of inflammation resolution. Based on findings from chapter 3, a biologically

active inflammation resolution program may exist followed exercise-induced muscle damage.

Future work is warranted to determine how this secondary wave of NF-κB pathway activity

influences post-exercise inflammation and skeletal muscle recovery.

Supplementation with the NSAID ibuprofen was used to investigate whether manipulating the

inflammatory response to exercise through the cyclooxygenase-prostaglandin pathway alters

81

post-exercise NF-κB signaling. Previous reports indicate that NF-κB can function upstream of

COX-2 to control transcription of this gene [192, 297]. Alternatively, prostaglandin activity may

also affect NF-κB [297-299]. In-vitro studies demonstrate that the effect of prostaglandins on

NF-κB activity is specific to the class of prostaglandin. Prostaglandin E2 activates the classical

NF-κB pathway, whereas prostaglandin A2, and prostaglandin J2 and its downstream analogues

can inhibit NF-κB activation in response to pro-inflammatory stimuli [298-301]. Our group

recently reported changes in prostaglandins in blood serum samples after traditional resistance

exercise [102]. Interestingly, PGE2, PGA2 and PGD2 all peaked in expression at 2 h post-exercise.

No significant time group interaction effects for the change in p65 phosphorylation was

observed; however, LSD comparisons suggested that phosphorylated p65 expression only

seemed to increase in the placebo group. Similarly, the observed changes in p50 protein

expression and DNA binding were only identified within the placebo group. These findings are

supported by data from the study by Lawrence et al [283], which demonstrated no change in

the secondary wave of NF-B DNA binding activity following the administration of an

alternative COX inhibitor, NS398, in rats with pleurisy. While these findings do not offer any

conclusive evidence that ibuprofen treatment inhibits the NF-κB pathway, it does provide

justification to further explore this pathway as a potential mechanism through which ibuprofen

treatment inhibits post-exercise inflammation.

4.6 Conclusion:

This research provides new insights into the regulation of NF-κB following a bout of acute

resistance exercise. The primary finding from this study is that NF-κB activation follows a

biphasic activation pattern that is subunit-specific. The first wave involves the classical NF-κB

p65 subunit, and corresponds with the onset of an acute inflammatory response and an

increase in inflammatory cytokine gene expression. The second wave is consistent with a

previously identified secondary wave of NF-κB activity that involves the alternative p50

subunit. Research in alternative models of inflammation suggests that this second wave of

activity may be associated with an active inflammatory resolution program. Further research

into the role of p50 signaling in skeletal muscle represents a key area for future research in

order to better understand the mechanisms that regulate post-exercise inflammation. Analysis

of the LSD to examine pair-wise within-group differences suggested that the observed changes

82

in both NF-κB p65 and p50 were detected only within the placebo group. The complex

interplay between the NF-κB and the COX/prostaglandin pathway in exercise-induced muscle

damage remains poorly understood, and should be a focus for future research.

83

CHAPTER 5 – GENERAL DISCUSSION AND FUTURE DIRECTIONS:

5.1 – Introduction:

The human response to resistance exercise presents a highly complex paradigm. Intense

muscular contractions have the capacity to trigger large increases in muscle size and strength.

However, following most bouts of intense exercise, there is often considerable muscle damage,

inflammation and muscle soreness. Whilst recognising the complexity of the skeletal muscle

response to exercise-induced muscle damage, the time-course of cellular events in post-

exercise muscle recovery appear to follow a well described schematic of events; (1) tissue

injury via mechanical and metabolic stress, (2) release of vasoactive and chemotactic

substances from damaged tissue, (3) migration and adhesion of systemic leucocytes to the

injury site, and (4) tissue repair and adaptation. The accumulation of inflammatory leucocytes

coincides with a secondary rise in histological and functional markers of muscle damage

including DOMS and a reduction in force generating capacity [24, 31, 39, 42, 302, 303]. These

findings have led researchers to propose that exercise-induced inflammation is a potential

cause of secondary muscle damage, and led to the implementation of anti-inflammatory

recovery strategies. However, with the past few years has been a growing recognition that

interventions targeted at the inhibition of key pro-inflammatory mediator pathways, have also

had deleterious effects of tissue adaptation and skeletal muscle repair [103, 106, 107, 161,

255]. These findings rationalize the concept that cellular events occurring within acute

inflammation form a key regulatory component of muscle regeneration and tissue repair.

The overall aim of my PhD was to explore the possibility of an active post-exercise

inflammatory resolution programme that may play a key role in establishing a biological link

between skeletal muscle damage and tissue repair. Specifically, chapter 2 involved a clinical

trial looking at the effect of ant-inflammatory medication on key markers of inflammation of

muscle damage. This project was part of a wider trial that examined the effect of anti-

inflammatory medication of muscle hypertrophy signaling pathways. The aim of chapter 3 was

to explore the human lipid profile in response to an acute resistance exercise. This paper

identified various lipids involved in regulating the bioactive resolution of acute inflammation.

These findings permitted the investigation of potential pathways that regulate both the

84

activation and resolution of acute inflammation. Chapter 4 examined the effect of an acute

bout of resistance exercise on the classical and alternative NF-κB signaling pathways in skeletal

muscle of young untrained men.

5.2 – Summary of results:

The aim for chapter 2 was to investigate the effect of oral ibuprofen administration on

leucocyte infiltration and indirect markers of muscle damage, including circulating muscle

proteins CK and myoglobin, and subjective muscle soreness. This was the first study to

demonstrate that ibuprofen administration has no effect on the histological appearance of

inflammatory white blood cells following an acute bout of traditional resistance exercise. We

also found no effect of ibuprofen administration on blood markers of muscle damage or

subjective muscle soreness, and no significant correlations between leucocyte numbers and

post-exercise muscle damage or soreness.

Chapter 3 sought to explore the possibly of a biologically active inflammatory resolution

pathway that may provide a physiological link between processes of acute exercise-induced

inflammation and skeletal muscle adaptation. Post-exercise recovery was characterized by

increased levels of COX-1 derived TXB2 and 12(S)-HHTrE, COX-2 derived prostaglandin

metabolites 2,3-dinor PGE1 and 15d-D12,14-PGJ3, LOX derivatives LTB4, 20-COOH LTB4, 5-HETE,

12-HETE, tetranor 12-HETE and 15-HETE, HDoHE species 4-HDoHE, 7-HDoHE, and CYP derived

5,6-EpETrE, 11,12-DiHETrE and 14,15-DiHETrE. This lipid profile suggests that novel lipid

intermediates involved in the formation of specialized pro-resolution mediators form a highly

complex self-resolving model of acute inflammation following a bout of resistance exercise.

The aim of chapter 4 was to explore the regulation of NF-κB as a potential regulator of an

active post-exercise inflammatory resolution programme. The primary finding from this study

is that NF-B activation follows a biphasic activation pattern that is subunit-specific. The first

wave involves the classical NF-B p65 subunit, and corresponds with the onset of an acute

inflammatory response and an increase in inflammatory cytokine gene expression. The second

wave is consistent with a previously identified secondary wave of NF-B activity that involves

the alternative p50 subunit. These findings are consistent with those from alternative models

85

of acute inflammation and identifies NF-κB as a critically important regulator of post-exercise

inflammation.

5.3 – Future directions:

This thesis represents a broad approach to exploring the possibility of a bioactive inflammatory

resolution programme following exercise-induced muscle damage. The important outcomes

from this research are; (1) leucocyte infiltration occurs following an acute resistance exercise

stimulus, however does not appear to be associated with exercise-induced muscle soreness,

(2) pro-resolution inflammatory mediators derived from omega-3 and omega-6 fatty acids are

increased following a bout of resistance exercise, (3) the biphasic regulation of NF-κB, a key

regulator of stress signaling pathways, coincides with the onset and resolution of acute

inflammation. Collectively, these findings provide preliminary evidence for an active

inflammatory resolution programme that may provide a biological link between processes of

acute muscle damage, inflammation and soft-tissue regeneration.

In chapter 2 I identified that the administration of a non-selective NSAID had no effect on the

observed increase in white blood cell infiltration or indirect measures of muscle damage.

Paulsen et al. (2010) proposed the concept of an inflammatory threshold, suggesting that

NSAIDs may only influence post-exercise inflammation in response to a sufficiently strong

inflammatory stimulus [93]. It was suggested that the absence of an intramuscular

prostaglandin response to exercise, specifically PGE2, could explain the lack of an NSAID effect

on leucocyte recruitment [93, 132]. Interestingly, in a follow up study within the same sample,

NSAID administration blunted early translational pathways involved in skeletal muscle

hypertrophy [161]. These findings present two potential paradigms. Firstly, despite the well-

established role for the inducible COX-2 enzyme in the animal muscle reparative response to

acute damage [104-106, 111, 164], the constitutively active COX-1 may play an important

function in the human adaptive response to exercise. Alternatively, NSAID administration may

have the capacity to influence alternative signalling pathways within the exercise-induced

inflammation-tissue repair paradigm.

86

These findings led our research team to explore the regulation of a novel family of n-3 and n-6

derived lipid mediators known to play a role in acute inflammatory resolution. In addition to

their role in regulating inflammation, lipid mediators have been implicated in processes of soft

tissue regeneration and skeletal muscle adaptation [304]. Firstly, Markworth et al. [102]

demonstrated elevated levels of pro-resolving lipoxins, resolvins and protectins in human

blood serum samples during the early hours of post-exercise muscle recovery. Treatment with

ibuprofen inhibited the lipid mediator response to exercise-induced cellular stress. In chapter 3

I identified a similar increase in lipids derived from the COX, LOX and CYP pathways in skeletal

muscle tissue following acute resistance exercise. In each research trial a unique lipid profile

was identified in blood serum and skeletal muscle tissue samples. Future research exploring

the functions of these lipids in exercise-induced muscle damage is necessary in understanding

the physiological significance of acute inflammation in post-exercise skeletal muscle

regeneration. At this stage the mechanism of action for lipid derived resolution mediators is

not clear and requires further investigation. This can be explored further in-vivo using rodent

models of muscle damage. Utilizing an eccentrically loaded muscle contraction, or an

unloading/reloading muscle damaging protocol, lipoxin and resolvin species can be artificially

over-expressed, or pharmacologically inhibited [171, 305, 306]. This research needs to consider

the implications of modifying lipid mediator synthesis on the onset and resolution of acute

inflammation, and what impact this has on downstream markers of skeletal muscle tissue

regeneration. Further human trials need to consider the effect of dietary interventions on key

markers of post-exercise muscle damage, inflammation and repair. Supplementation with n-3

EPA and DPA has a highly divergent effect on the human lipid mediator profile, specifically

resolvin and maresin species [307]. This avenue of research would explore the concept of

recovery interventions designed to promote resolving mediators and biochemical pathways

that are homeostatic and modulatory in their actions, to optimize the outcome of an acute

inflammatory program.

Chapter 4 identified NF-κB as a potential transcriptional regulator involved in both the onset

and resolution of acute post-exercise inflammation. The primary finding from this study is that

NF-κB regulation follows a biphasic activation pattern that is subunit-specific. The first wave

involves the classical NF-κB p65 subunit, and corresponds with the onset of an acute

87

inflammatory response and an increase in inflammatory cytokine gene expression. The second

wave is consistent with a previously identified secondary wave of NF-κB activity that involves

the alternative p50 subunit. While research in alternative models of acute inflammation

suggest this second wave of increased NF-κB activity may be associated with an active

inflammatory resolution program, no research has explored the physiological significance of

this pathway in acute post-exercise inflammation. Recent work from our group utilized a series

of electrophoretic mobility shift assays (EMSA) to demonstrate an increase in NF-κB p65

binding to the promoter region of genes coding for inflammatory cytokines in the early stages

of exercise-induced inflammation [239]. This research model may provide an avenue to explore

whether the secondary increase in key markers of the alternative NF-κB signaling pathway are

involved in regulating a biologically active inflammatory resolution program. Potential targets

of NF-κB p50 homodimers include the inducible COX-2 enzyme, transcription factor p53 and

the pro-apoptotic Bcl2 homolog Bax [192, 210].

An inherent limitation of muscle biopsy studies is the test-retest reliability of muscle biopsies

obtained from different sites along the vastus lateralis. The muscle biopsy protocol used

throughout this thesis was used to minimize interference from the biopsy procedure on

markers of acute inflammation [239]. However, previous research has demonstrated the

regional differences in muscle hypertrophy/atrophy exist across an individual muscle length

[308]. Further evidence suggests that intramuscular differences in fibre type have been

observed across individual muscle biopsy samples from the vastus lateralis [309]. The absence

of a non-exercise control group, and the inherent lack of test-retest reliability of the muscle

biopsy protocol are acknowledged as accepted limitations of the current research.

5.4 – Concluding remarks:

This thesis presents preliminary evidence for a biologically active inflammatory resolution

program that may provide a crucial link between processes of exercise-induced muscle

damage, and tissue regeneration and adaptation. This concept would challenge the current use

of anti-inflammatory therapies to promote post-exercise muscle recovery. Most of the anti-

inflammatory interventions studied to date, arise from our need to control the cardinal signs of

88

inflammation by inhibiting key pro-inflammatory mediator pathways. Experimental models

targeted at inhibiting the post-exercise inflammatory response have identified that exercise-

induced inflammation is a key regulatory feature in the restoration of tissue structure and

function.

The overall aim of this thesis was to explore the regulation of a family of novel lipid derivatives

that have been shown to play dual anti-inflammatory and pro-resolution functions in acute

inflammation. In chapter 2 we demonstrated that the administration of ibuprofen had no

effect on key inflammatory and muscle damage markers. These findings challenge the

suitability of NSAID administration as an effective treatment strategy for reducing acute

inflammation and muscle soreness following resistance exercise. Further research from within

our laboratory [161], amongst others [87, 107, 130], have demonstrated a detrimental effect

of anti-inflammatory interventions on post-exercise early translational signalling responses via

the inhibition of eicosanoids derived from the COX pathway. Taken together, these findings

suggest that inhibiting eicosanoid formation during acute post-exercise inflammation may

directly affect muscle hypertrophy pathways without directly impacting on cellular

inflammation. In chapter 3 we explored the regulation of lipid species during acute post-

exercise muscle recovery. We identified a post-exercise increase in eicosanoids and

docosanoids species derived from COX, LOX and CYP pathways, presenting the possibility of a

highly complex self-resolving model of acute inflammation following an acute bout of

resistance exercise. For chapter 4, an in-depth analysis of the regulation of NF-κB,

demonstrated a bi-phasic regulation pattern that coincides with the onset and resolution of

acute inflammation. This thesis shows for the first time that events occurring early in acute

post-exercise inflammation may engage an active and coordinated inflammatory resolution

programme, governed by the production of specialized pro-resolution lipid mediators. Further

work is required to elucidate the specific functions of each of these lipid variables and explore

the efficacy of interventions designed to promote resolving mediators and biochemical

pathways that will be homeostatic and modulatory in their actions.

89

REFERENCE LIST:

90

1. Flück, M., Functional, structural and molecular plasticity of mammalian skeletal muscle

in response to exercise stimuli. The Journal Of Experimental Biology, 2006. 209(Pt 12): p.

2239-2248.

2. Harridge, S.D.R., Plasticity of human skeletal muscle: gene expression to in vivo function.

Experimental Physiology, 2007. 92(5): p. 783-797.

3. Rasmussen, B.B. and S.M. Phillips, Contractile and nutritional regulation of human

muscle growth. Exercise And Sport Sciences Reviews, 2003. 31(3): p. 127-131.

4. Schiaffino, S. and G. Salviati, Molecular diversity of myofibrillar proteins: isoforms

analysis at the protein and mRNA level. Methods In Cell Biology, 1997. 52: p. 349-369.

5. Seynnes, O.R., M. de Boer, and M.V. Narici, Early skeletal muscle hypertrophy and

architectural changes in response to high-intensity resistance training. Journal of

Applied Physiology, 2007. 102(1): p. 368-373.

6. Vierck, J., et al., Satellite cell regulation following myotrauma caused by resistance

exercise. Cell Biology International, 2000. 24(5): p. 263-272.

7. Hough, T., Ergographic studies in muscular fatigue and soreness. Journal. Boston

Society Of Medical Sciences, 1900. 5(3): p. 81-92.

8. Faulkner, J.A., D.A. Jones, and J.M. Round, Injury to skeletal muscles of mice by forced

lengthening during contractions. Quarterly Journal Of Experimental Physiology

(Cambridge, England), 1989. 74(5): p. 661-670.

9. Armstrong, R.B., R.W. Ogilvie, and J.A. Schwane, Eccentric exercise-induced injury to rat

skeletal muscle. Journal Of Applied Physiology: Respiratory, Environmental And Exercise

Physiology, 1983. 54(1): p. 80-93.

10. Kuipers, H., et al., Muscle degeneration after exercise in rats. International Journal Of

Sports Medicine, 1983. 4(1): p. 45-51.

11. Lieber, R.L., L.E. Thornell, and J. Fridén, Muscle cytoskeletal disruption occurs within the

first 15 min of cyclic eccentric contraction. Journal Of Applied Physiology (Bethesda,

Md.: 1985), 1996. 80(1): p. 278-284.

91

12. Lieber, R.L., T.M. Woodburn, and J. Fridén, Muscle damage induced by eccentric

contractions of 25% strain. Journal Of Applied Physiology (Bethesda, Md.: 1985), 1991.

70(6): p. 2498-2507.

13. McNeil, P.L. and R. Khakee, Disruptions of muscle fiber plasma membranes. Role in

exercise-induced damage. The American Journal Of Pathology, 1992. 140(5): p. 1097-

1109.

14. Salminen, A., K. Hongisto, and V. Vihko, Lysosomal changes related to exercise injuries

and training-induced protection in mouse skeletal muscle. Acta Physiologica

Scandinavica, 1984. 120(1): p. 15-19.

15. Salminen, A. and V. Vihko, Exercise myopathy: selectively enhanced proteolytic capacity

in rat skeletal muscle after prolonged running. Experimental And Molecular Pathology,

1983. 38(1): p. 61-68.

16. Fridén, J. and R.L. Lieber, Segmental muscle fiber lesions after repetitive eccentric

contractions. Cell And Tissue Research, 1998. 293(1): p. 165-171.

17. Fridén, J., M. Sjöström, and B. Ekblom, A morphological study of delayed muscle

soreness. Experientia, 1981. 37(5): p. 506-507.

18. Fridén, J., U. Kjörell, and L.E. Thornell, Delayed muscle soreness and cytoskeletal

alterations: an immunocytological study in man. International Journal Of Sports

Medicine, 1984. 5(1): p. 15-18.

19. Friden, J., M. Sjoestroem, and B. Ekblom, Myofibrillar damage following intense

eccentric exercise in man. International Journal of Sports Medicine, 1983. 4(3): p. 170-

176.

20. Newham, D.J., The consequences of eccentric contractions and their relationship to

delayed onset muscle pain. European Journal Of Applied Physiology And Occupational

Physiology, 1988. 57(3): p. 353-359.

21. Newham, D.J., D.A. Jones, and R.H.T. Edwards, Large delayed plasma creatine kinase

changes after stepping exercise. Muscle & Nerve, 1983(6): p. 380-385.

92

22. Newham, D.J., et al., Muscle fatigue and pain after eccentric contractions at long and

short length. Clinical Science (London, England: 1979), 1988. 74(5): p. 553-557.

23. Pillon, N.J., et al., Cross-talk between skeletal muscle and immune cells: muscle-derived

mediators and metabolic implications. American Journal of Physiology - Endocrinology

and Metabolism, 2013. 304(5): p. E453-E465.

24. Armstrong, R.B., G.L. Warren, and J.A. Warren, Mechanisms of exercise-induced muscle

fibre injury. Sports Medicine, 1991. 12(3): p. 184-207.

25. Smith, L.L., Acute inflammation: the underlying mechanism in delayed onset muscle

soreness? Medicine & Science in Sports & Exercise, 1991. 23(5): p. 542-551.

26. Appell, H.J., J.M.C. Soares, and J.A.R. Duarte, Exercise, muscle damage and fatigue.

Sports Medicine, 1992. 13(2): p. 108-115.

27. Lieber, R.L. and J. Fridén, Mechanisms of muscle injury gleaned from animal models.

American Journal Of Physical Medicine & Rehabilitation / Association Of Academic

Physiatrists, 2002. 81(11 Suppl): p. S70-S79.

28. Michna, H., Ultrastructural features of skeletal muscle in mice after physical exercise: its

relation to the pathogenesis of leucocyte invasion. Acta Anatomica, 1989. 134(4): p.

276-282.

29. Smith, C., et al., The inflammatory response to skeletal muscle injury: illuminating

complexities. Sports Medicine (Auckland, N.Z.), 2008. 38(11): p. 947-969.

30. Tidball, J.G., Inflammatory processes in muscle injury and repair. American Journal Of

Physiology. Regulatory, Integrative And Comparative Physiology, 2005. 288(2): p. R345-

R353.

31. Tidball, J.G., Inflammatory cell response to acute muscle injury. Medicine & Science in

Sports & Exercise, 1995. 27(7): p. 1022-1032.

32. Stauber, W.T., et al., Extracellular matrix disruption and pain after eccentric muscle

action. Journal of Applied Physiology, 1990. 69(3): p. 868-874.

93

33. Tidball, J.G. and S.A. Villalta, Regulatory interactions between muscle and the immune

system during muscle regeneration. American Journal Of Physiology. Regulatory,

Integrative And Comparative Physiology, 2010. 298(5): p. R1173-R1187.

34. Butterfield, T.A., T.M. Best, and M.A. Merrick, The Dual Roles of Neutrophils and

Macrophages in Inflammation: A Critical Balance Between Tissue Damage and Repair.

Journal of Athletic Training, 2006. 41(4): p. 457-465.

35. Evans, W.J. and J.G. Cannon, The metabolic effects of exercise-induced muscle damage.

Exercise & Sport Sciences Reviews, 1991. 19: p. 99-125.

36. Frenette, J., et al., Muscle impairment occurs rapidly and precedes inflammatory cell

accumulation after mechanical loading. American Journal of Physiology: Regulatory,

Integrative & Comparative Physiology, 2002. 51(2): p. R351.

37. Nguyen, H.X. and J.G. Tidball, Null mutation of gp91phox reduces muscle membrane

lysis during muscle inflammation in mice. The Journal Of Physiology, 2003. 553(Pt 3): p.

833-841.

38. Toumi, H., S. F’guyer, and T.M. Best, The role of neutrophils in injury and repair

following muscle stretch. Journal of Anatomy, 2006. 208(4): p. 459-470.

39. Brickson, S., et al., Oxidant production and immune response after stretch injury in

skeletal muscle. Medicine & Science in Sports & Exercise, 2001. 33(12): p. 2010-2015.

40. Hurme, T., et al., Healing of skeletal muscle injury: an ultrastructural and

immunohistochemical study. Medicine & Science in Sports & Exercise, 1991. 23(7): p.

801-810.

41. Newham, D.J., et al., Ultrastructural changes after concentric and eccentric contractions

of human muscle. Journal of Neurological Sciences, 1983. 61(1): p. 109-122.

42. Pizza, F.X., et al., Neutrophils contribute to muscle injury and impair its resolution after

lengthening contractions in mice. The Journal Of Physiology, 2005. 562(Pt 3): p. 899-

913.

43. St Pierre Schneider, B., et al., CD11b+ neutrophils predominate over RAM11+

macrophages in stretch-injured muscle. Muscle & Nerve, 2002. 25(6): p. 837-844.

94

44. Erwig, L.P., et al., Initial cytokine exposure determines function of macrophages and

renders them unresponsive to other cytokines. Journal Of Immunology (Baltimore, Md.:

1950), 1998. 161(4): p. 1983-1988.

45. MacIntyre, D.L., W.D. Reid, and D.C. McKenzie, Delayed muscle soreness. The

inflammatory response to muscle injury and its clinical implications. Sports Medicine

(Auckland, N.Z.), 1995. 20(1): p. 24-40.

46. Honda, H., H. Kimura, and A. Rostami, Demonstration and phenotypic characterization

of resident macrophages in rat skeletal muscle. Immunology, 1990. 70(2): p. 272-277.

47. St. Pierre, B.A. and J.G. Tidball, Differential response of macrophage subpopulations to

soleus muscle reloading after rat hindlimb suspension. Journal of Applied Physiology,

1994. 77(1): p. 290-297.

48. Arnold, L., et al., Inflammatory monocytes recruited after skeletal muscle injury switch

into antiinflammatory macrophages to support myogenesis. The Journal Of

Experimental Medicine, 2007. 204(5): p. 1057-1069.

49. McLennan, I.S., Degenerating and regenerating skeletal muscles contain several

subpopulations of macrophages with distinct spatial and temporal distributions. Journal

of Anatomy, 1996. 188 ( Pt 1): p. 17-28.

50. Krippendorf, B.B., Riley, D. A., Distinguishing unloading versus reloading-induced

changes in rat soleus muscle. Muscle & Nerve, 1993. 16: p. 99-108.

51. McLennan, I.S., Resident macrophages (ED2- and ED3-positive) do not phagocytose

degenerating rat skeletal muscle fibres. Cell And Tissue Research, 1993. 272(1): p. 193-

196.

52. Honda, H., H. Kimura, and A. Rostami, Isolation and characterization of macrophages

from rat embryonic muscle culture. Journal Of Leukocyte Biology, 1992. 52(5): p. 537-

544.

53. Cantini, M., et al., Macrophage-secreted myogenic factors: a promising tool for greatly

enhancing the proliferative capacity of myoblasts in vitro and in vivo. Neurological

Sciences, 2002. 23(4): p. 189.

95

54. Chazaud, B., et al., Satellite cells attract monocytes and use macrophages as a support

to escape apoptosis and enhance muscle growth. Journal of Cell Biology, 2003. 163(5):

p. 1133-1143.

55. Gierer, P., et al., Selective cyclooxygenase-2 inhibition reverses microcirculatory and

inflammatory sequelae of closed soft tissue trauma in an animal model. Journal of Bone

& Joint Surgery, American Volume, 2005. 87(1): p. 153-160.

56. Massimino, M.L., et al., ED2+ macrophages increase selectively myoblast proliferation in

muscle cultures. Biochemical And Biophysical Research Communications, 1997. 235(3):

p. 754-759.

57. Merly, F., et al., Macrophages enhance muscle satellite cell proliferation and delay their

differentiation. Muscle & Nerve, 1999. 22(6): p. 724-732.

58. Sonnet, C., et al., Human macrophages rescue myoblasts and myotubes from apoptosis

through a set of adhesion molecular systems. Journal Of Cell Science, 2006. 119(Pt 12):

p. 2497-2507.

59. Summan, M., et al., Macrophages and skeletal muscle regeneration: a clodronate-

containing liposome depletion study. American Journal Of Physiology. Regulatory,

Integrative And Comparative Physiology, 2006. 290(6): p. R1488-R1495.

60. Tidball, J.G. and M. Wehling-Henricks, Macrophages promote muscle membrane repair

and muscle fibre growth and regeneration during modified muscle loading in mice in

vivo. The Journal Of Physiology, 2007. 578(Pt 1): p. 327-336.

61. Crameri, R.M., et al., Myofibre damage in human skeletal muscle: effects of electrical

stimulation versus voluntary contraction. The Journal Of Physiology, 2007. 583(Pt 1): p.

365-380.

62. Malm, C., et al., Immunological changes in human skeletal muscle and blood after

eccentric exercise and multiple biopsies. The Journal Of Physiology, 2000. 529 Pt 1: p.

243-262.

96

63. Raastad, T., et al., Temporal relation between leukocyte accumulation in muscles and

halted recovery 10-20 h after strength exercise. Journal Of Applied Physiology

(Bethesda, Md.: 1985), 2003. 95(6): p. 2503-2509.

64. Yu, J.-G., D.O. Fürst, and L.-E. Thornell, The mode of myofibril remodelling in human

skeletal muscle affected by DOMS induced by eccentric contractions. Histochemistry

And Cell Biology, 2003. 119(5): p. 383-393.

65. Yu, J.-G., C. Malm, and L.-E. Thornell, Eccentric contractions leading to DOMS do not

cause loss of desmin nor fibre necrosis in human muscle. Histochemistry and Cell

Biology, 2002. 118(1): p. 29-34.

66. MacIntyre, D.L., et al., Presence of WBC, decreased strength, and delayed soreness in

muscle after eccentric exercise. Journal Of Applied Physiology (Bethesda, Md.: 1985),

1996. 80(3): p. 1006-1013.

67. Crenshaw, A.G., et al., Increased technetium uptake is not equivalent to muscle necrosis:

scintigraphic, morphological and intramuscular pressure analyses of sore muscles after

exercise. Acta Physiologica Scandinavica, 1993. 148(2): p. 187-198.

68. Fielding, R.A., et al., Acute phase response in exercise. III. Neutrophil and IL-1 beta

accumulation in skeletal muscle. The American Journal Of Physiology, 1993. 265(1 Pt 2):

p. R166-R172.

69. Hellsten, Y., et al., Xanthine oxidase in human skeletal muscle following eccentric

exercise: a role in inflammation. The Journal Of Physiology, 1997. 498 ( Pt 1): p. 239-

248.

70. Jones, D.A., et al., Experimental human muscle damage: morphological changes in

relation to other indices of damage. The Journal Of Physiology, 1986. 375: p. 435-448.

71. Kuipers, H., et al., Influence of a prostaglandin-inhibiting drug on muscle soreness after

eccentric work. International Journal Of Sports Medicine, 1985. 6(6): p. 336-339.

72. Nurenberg, P., et al., MR imaging-guided muscle biopsy for correlation of increased

signal intensity with ultrastructural change and delayed-onset muscle soreness after

exercise. Radiology, 1992. 184(3): p. 865-869.

97

73. O'Reilly, K.P., et al., Eccentric exercise-induced muscle damage impairs muscle glycogen

repletion. Journal Of Applied Physiology (Bethesda, Md.: 1985), 1987. 63(1): p. 252-256.

74. Round, J.M., D.A. Jones, and G. Cambridge, Cellular infiltrates in human skeletal muscle:

exercise induced damage as a model for inflammatory muscle disease? Journal Of The

Neurological Sciences, 1987. 82(1-3): p. 1-11.

75. Aronson, D., et al., Extracellular-regulated protein kinase cascades are activated in

response to injury in human skeletal muscle. The American Journal Of Physiology, 1998.

275(2 Pt 1): p. C555-C561.

76. Constantin-Teodosiu, D., et al., The effect of repeated muscle biopsy sampling on ATP

and glycogen resynthesis following exercise in man. European Journal Of Applied

Physiology And Occupational Physiology, 1996. 73(1-2): p. 186-190.

77. Chleboun, G.S., J.N. Howell, and R.R. Conatser, Relationship between muscle swelling

and stiffness after eccentric exercise. Medicine & Science in Sports & Exercise, 1998.

30(4): p. 529-535.

78. Cleak, M.J. and R.G. Eston, Muscle soreness, swelling, stiffness and strength loss after

intense eccentric exercise. British Journal Of Sports Medicine, 1992. 26(4): p. 267-272.

79. Fridén, J. and R.L. Lieber, Serum creatine kinase level is a poor predictor of muscle

function after injury. Scandinavian Journal Of Medicine & Science In Sports, 2001. 11(2):

p. 126-127.

80. Nosaka, K. and P.M. Clarkson, Variability in serum creatine kinase response after

eccentric exercise of the elbow flexors. International Journal Of Sports Medicine, 1996.

17(2): p. 120-127.

81. Nosaka, K., M. Newton, and P. Sacco, Delayed-onset muscle soreness does not reflect

the magnitude of eccentric exercise-induced muscle damage. Scandinavian Journal of

Medicine & Science in Sports, 2002. 12(6): p. 337.

82. Rodenburg, J., P. Bar, and R. De Baer, Relations between muscle soreness and

biochemical and functional outcomes of eccentric exercise. Journal of Applied

Physiology, 1993. 74(6): p. 2976-2983.

98

83. Féasson, L., et al., Molecular adaptations of neuromuscular disease-associated proteins

in response to eccentric exercise in human skeletal muscle. The Journal Of Physiology,

2002. 543(Pt 1): p. 297-306.

84. Paulsen, G., et al., Time course of leukocyte accumulation in human muscle after

eccentric exercise. Medicine And Science In Sports And Exercise, 2010. 42(1): p. 75-85.

85. Paulsen, G., et al., Leucocytes, cytokines and satellite cells: what role do they play in

muscle damage and regeneration following eccentric exercise? Exercise Immunology

Review, 2012. 18: p. 42-97.

86. Hubal, M.J., et al., Inflammatory gene changes associated with the repeated-bout

effect. American Journal of Physiology: Regulatory, Integrative & Comparative

Physiology, 2008. 63(5): p. R1628-R1637.

87. Mikkelsen, U.R., et al., Local NSAID infusion inhibits satellite cell proliferation in human

skeletal muscle after eccentric exercise. Journal Of Applied Physiology (Bethesda, Md.:

1985), 2009. 107(5): p. 1600-1611.

88. Beaton, L.J., et al., Contraction-induced muscle damage is unaffected by vitamin E

supplementation. Medicine And Science In Sports And Exercise, 2002. 34(5): p. 798-805.

89. Beaton, L.J., M.A. Tarnopolsky, and S.M. Phillips, Contraction-induced muscle damage in

humans following calcium channel blocker administration. The Journal Of Physiology,

2002. 544(Pt 3): p. 849-859.

90. Crenshaw, A.G., et al., Knee extension torque and intramuscular pressure of the vastus

lateralis muscle during eccentric and concentric activities. European Journal Of Applied

Physiology And Occupational Physiology, 1995. 70(1): p. 13-19.

91. Crenshaw, A.G., L.E. Thornell, and J. Fridén, Intramuscular pressure, torque and swelling

for the exercise-induced sore vastus lateralis muscle. Acta Physiologica Scandinavica,

1994. 152(3): p. 265-277.

92. Stupka, N., et al., Cellular adaptation to repeated eccentric exercise-induced muscle

damage. Journal Of Applied Physiology (Bethesda, Md.: 1985), 2001. 91(4): p. 1669-

1678.

99

93. Paulsen, G., et al., A COX-2 inhibitor reduces muscle soreness, but does not influence

recovery and adaptation after eccentric exercise. Scandinavian Journal of Medicine &

Science in Sports, 2010. 20(1): p. 1-13.

94. Child, R., et al., Changes in indices of antioxidant status, lipid peroxidation and

inflammation in human skeletal muscle after eccentric muscle actions. Clinical Science

(London, England: 1979), 1999. 96(1): p. 105-115.

95. Hikida, R.S., et al., Muscle fiber necrosis associated with human marathon runners.

Journal Of The Neurological Sciences, 1983. 59(2): p. 185-203.

96. Lauritzen, F., et al., Gross ultrastructural changes and necrotic fiber segments in elbow

flexor muscles after maximal voluntary eccentric action in humans. Journal of Applied

Physiology, 2009. 107(6): p. 1923-1934.

97. Paulsen, G., et al., Inflammatory markers CD11b, CD16, CD66b, CD68, myeloperoxidase

and neutrophil elastase in eccentric exercised human skeletal muscles. Histochemistry

And Cell Biology, 2013. 139(5): p. 691-715.

98. Mahoney, D.J., et al., Gene expression profiling in human skeletal muscle during

recovery from eccentric exercise. American Journal Of Physiology. Regulatory,

Integrative And Comparative Physiology, 2008. 294(6): p. R1901-R1910.

99. Elnachef, N., et al., Changing perceptions and practices regarding aspirin, nonsteroidal

anti-inflammatory drugs, and cyclooxygenase-2 selective nonsteroidal anti-

inflammatory drugs among US primary care providers. Alimentary Pharmacology &

Therapeutics, 2008. 28(10): p. 1249-1258.

100. Green, G.A., Understanding NSAIDs: from aspirin to COX-2. Clinical Cornerstone, 2001.

3(5): p. 50-60.

101. Markworth, J.F., K.R. Maddipati, and D. Cameron-Smith, Emerging roles of pro-resolving

lipid mediators in immunological and adaptive responses to exercise-induced muscle

injury. Exercise Immunology Review, 2016. 22: p. 110-134.

100

102. Markworth, J.F., et al., Human inflammatory and resolving lipid mediator responses to

resistance exercise and ibuprofen treatment. American Journal Of Physiology.

Regulatory, Integrative And Comparative Physiology, 2013. 305(11): p. R1281-R1296.

103. Urso, M.L., Anti-inflammatory interventions and skeletal muscle injury: benefit or

detriment? Journal Of Applied Physiology (Bethesda, Md.: 1985), 2013. 115(6): p. 920-

928.

104. Bondesen, B.A., et al., The COX-2 pathway is essential during early stages of skeletal

muscle regeneration. American Journal of Physiology: Cell Physiology, 2004. 56(2): p.

C475-C483.

105. Bondesen, B.A., S.T. Mills, and G.K. Paviath, The COX-2 pathway regulates growth of

atrophied muscle via multiple mechanisms. American Journal of Physiology: Cell

Physiology, 2006. 59(6): p. C1651-C1659.

106. Mishra, D.K., et al., Anti-inflammatory medication after muscle injury. A treatment

resulting in short-term improvement but subsequent loss of muscle function. The

Journal Of Bone And Joint Surgery. American Volume, 1995. 77(10): p. 1510-1519.

107. Trappe, T.A., et al., Effect of ibuprofen and acetaminophen on postexercise muscle

protein synthesis. American Journal Of Physiology. Endocrinology And Metabolism,

2002. 282(3): p. E551-E556.

108. Mackey, A.L., et al., The influence of anti-inflammatory medication on exercise-induced

myogenic precursor cell responses in humans. Journal of Applied Physiology, 2007.

103(2): p. 425-431.

109. Demers, L.M., et al., Effect of prolonged exercise on plasma prostaglandin levels.

Prostaglandins And Medicine, 1981. 6(4): p. 413-418.

110. Laustiola, K., et al., Exercise-induced increase in plasma arachidonic acid and

thromboxane B2 in healthy men: effect of beta-adrenergic blockade. Journal of

Cardiovascular Pharmacology, 1984. 6(3): p. 449-454.

101

111. Nowak, J. and A. Wennmalm, Effect of exercise on human arterial and regional venous

plasma concentrations of prostaglandin E. Prostaglandins And Medicine, 1978. 1(6): p.

489-497.

112. Venkatraman, J.T., X. Feng, and D. Pendergast, Effects of dietary fat and endurance

exercise on plasma cortisol, prostaglandin E2, interferon-gamma and lipid peroxides in

runners. Journal Of The American College Of Nutrition, 2001. 20(5): p. 529-536.

113. Carter, J.W., et al., The effect of exercise on bleeding time and local production of

prostacyclin and thromboxane. European Journal Of Applied Physiology And

Occupational Physiology, 1989. 59(5): p. 355-359.

114. Feng, D.L., et al., Upright posture and maximal exercise increase platelet aggregability

and prostacyclin production in healthy male subjects. British Journal Of Sports

Medicine, 1999. 33(6): p. 401-404.

115. Mourits-Andersen, T., et al., Plasma 6-keto-PGF1 alpha, thromboxane B2 and PGE2 in

type 1 (insulin-dependent) diabetic patients during exercise. Diabetologia, 1987. 30(7):

p. 460-463.

116. Vinikka, L., J. Vuori, and O. Ylikorkala, Lipid peroxides, prostacyclin, and thromboxane A2

in runners during acute exercise. Medicine & Science in Sports & Exercise, 1984. 16(3):

p. 275-277.

117. Dousset, E., et al., Bimodal recovery pattern in human skeletal muscle induced by

exhaustive stretch-shortening cycle exercise. Medicine And Science In Sports And

Exercise, 2007. 39(3): p. 453-460.

118. Uchida, M.C., et al., Effect of bench press exercise intensity on muscle soreness and

inflammatory mediators. Journal of Sports Sciences, 2009. 27(5): p. 499-507.

119. Tartibian, B., B.H. Maleki, and A. Abbasi, Omega-3 Fatty Acids Supplementation

Attenuates Inflammatory Markers After Eccentric Exercise in Untrained Men. Clinical

Journal of Sport Medicine, 2011. 21(2): p. 131-137.

102

120. Boatwright, D., R. Byrd, and M. Mangum, Relationship between serum prostaglandin

formation, creatine kinase activity, and ratings of perceived soreness. Research in

Sports Medicine: An International Journal, 1991. 2(2): p. 85-88.

121. Croisier, J.L., et al., Myocellular Enzyme Leakage, Polymorphonuclear Neutrophil

Activation and Delayed Onset Muscle Soreness Induced by Isokinetic Eccentric Exercise.

Archives of Physiology and Biochemistry, 1996. 104(3): p. 322-329.

122. Croisier, J.L., et al., Piroxicam fails to reduce myocellular enzyme leakage and delayed

onset muscle soreness induced by isokinetic eccentric exercise. Mediators Of

Inflammation, 1996. 5(3): p. 230-234.

123. Black, C.D., et al., Ginger (Zingiber officinale) reduces muscle pain caused by eccentric

exercise. Journal of Pain, 2010. 11(9): p. 894-903.

124. Braun, W.A., et al., The effects of chondroitin sulfate supplementation on indices of

muscle damage induced by eccentric arm exercise. The Journal Of Sports Medicine And

Physical Fitness, 2005. 45(4): p. 553-560.

125. Hirose, L., et al., Changes in inflammatory mediators following eccentric exercise of the

elbow flexors. Exercise Immunology Review, 2004. 10: p. 75-90.

126. Buford, T.W., et al., Protease supplementation improves muscle function after eccentric

exercise. Medicine & Science in Sports & Exercise, 2009. 41(10): p. 1908-1914.

127. Carroll, C.C., et al., The influence of acute resistance exercise on cyclooxygenase-1 and -

2 activity and protein levels in human skeletal muscle. American Journal Of Physiology.

Regulatory, Integrative And Comparative Physiology, 2013. 305(1): p. R24-R30.

128. Testa, M., et al., Expression and activity of cyclooxygenase isoforms in skeletal muscles

and myocardium of humans and rodents. Journal Of Applied Physiology (Bethesda, Md.:

1985), 2007. 103(4): p. 1412-1418.

129. Weinheimer, E.M., et al., Resistance exercise and cyclooxygenase (COX) expression in

human skeletal muscle: implications for COX-inhibiting drugs and protein synthesis.

American Journal Of Physiology. Regulatory, Integrative And Comparative Physiology,

2007. 292(6): p. R2241-R2248.

103

130. Burd, N.A., et al., Effect of a cyclooxygenase-2 inhibitor on postexercise muscle protein

synthesis in humans. American Journal of Physiology: Endocrinology & Metabolism,

2010. 61(2): p. E354-E361.

131. Trappe, T.A., et al., Influence of acetaminophen and ibuprofen on skeletal muscle

adaptations to resistance exercise in older adults. American Journal Of Physiology.

Regulatory, Integrative And Comparative Physiology, 2011. 300(3): p. R655-R662.

132. Trappe, T.A., et al., Skeletal muscle PGF(2)(alpha) and PGE(2) in response to eccentric

resistance exercise: influence of ibuprofen acetaminophen. The Journal Of Clinical

Endocrinology And Metabolism, 2001. 86(10): p. 5067-5070.

133. Trappe, T., et al., Effects of age and resistance exercise on skeletal muscle interstitial

prostaglandin F(2alpha). Prostaglandins, Leukotrienes, And Essential Fatty Acids, 2006.

74(3): p. 175-181.

134. Karamouzis, M., et al., The response of muscle interstitial prostaglandin E(2)(PGE(2)),

prostacyclin I(2)(PGI(2)) and thromboxane A(2)(TXA(2)) levels during incremental

dynamic exercise in humans determined by in vivo microdialysis. Prostaglandins,

Leukotrienes, And Essential Fatty Acids, 2001. 64(4-5): p. 259-263.

135. Karamouzis, M., et al., In situ microdialysis of intramuscular prostaglandin and

thromboxane in contracting skeletal muscle in humans. Acta Physiologica Scandinavica,

2001. 171(1): p. 71-76.

136. Boushel, R., et al., Combined inhibition of nitric oxide and prostaglandins reduces

human skeletal muscle blood flow during exercise. The Journal Of Physiology, 2002.

543(Pt 2): p. 691-698.

137. Burian, A., et al., An exploratory microdialysis study investigating the effect of repeated

application of a diclofenac epolamine medicated plaster on prostaglandin

concentrations in skeletal muscle after standardized physical exercise. British Journal Of

Clinical Pharmacology, 2013. 76(6): p. 880-887.

138. Dudley, G.A., et al., Efficacy of naproxen sodium for exercise-induced dysfunction muscle

injury and soreness. Clinical Journal Of Sport Medicine: Official Journal Of The Canadian

Academy Of Sport Medicine, 1997. 7(1): p. 3-10.

104

139. Reynolds, J.F., et al., Non-steroidal anti-inflammatory drugs fail to enhance healing of

acute hamstring injuries treated with physiotherapy. South African Medical Journal,

1995. 85(6): p. 517-522.

140. Tokmakidis, S.P., et al., The effects of ibuprofen on delayed muscle soreness and

muscular performance after eccentric exercise. Journal Of Strength And Conditioning

Research / National Strength & Conditioning Association, 2003. 17(1): p. 53-59.

141. Donnelly, A.W., R.J. Maughan, and P.H. Whiting, Effects of ibuprofen on exercise-

induced muscle soreness and indices of muscle damage. British Journal of Sports

Medicine, 1990. 24(3): p. 191-195.

142. Hasson, S.M., et al., Effect of ibuprofen use on muscle soreness, damage, and

performance: a preliminary investigation. Medicine & Science in Sports & Exercise,

1993. 25(1): p. 9-17.

143. Hill, D.W. and J.D. Richardson, Effectiveness of 10% trolamine salicylate cream on

muscular soreness induced by a reproducible program of weight training. The Journal Of

Orthopaedic And Sports Physical Therapy, 1989. 11(1): p. 19-23.

144. Lecomte, J.M., V.J. Lacroix, and D.L. Montgomery, A randomized controlled trial of the

effect of naproxen on delayed onset muscle soreness and muscle strength. Clinical

Journal Of Sport Medicine: Official Journal Of The Canadian Academy Of Sport

Medicine, 1998. 8(2): p. 82-87.

145. O'Grady, M., et al., Diclofenac sodium (Voltaren) reduced exercise-induced injury in

human skeletal muscle. Medicine & Science in Sports & Exercise, 2000. 32(7): p. 1191-

1196.

146. Rahnama, N., F. Rahmani-Nia, and K. Ebrahim, The isolated and combined effects of

selected physical activity and ibuprofen on delayed-onset muscle soreness. Journal Of

Sports Sciences, 2005. 23(8): p. 843-850.

147. Bourgeois, J., et al., Naproxen does not alter indices of muscle damage in resistance-

exercise trained men. Medicine & Science in Sports & Exercise, 1999. 31(1): p. 4-9.

105

148. Baldwin, A.C., S.W. Stevenson, and G.A. Dudley, Nonsteroidal anti-inflammatory

therapy after eccentric exercise in healthy older individuals. The Journals Of

Gerontology. Series A, Biological Sciences And Medical Sciences, 2001. 56(8): p. M510-

M513.

149. Cannavino, C.R., et al., Efficacy of transdermal ketoprofen for delayed onset muscle

soreness. Clinical Journal Of Sport Medicine: Official Journal Of The Canadian Academy

Of Sport Medicine, 2003. 13(4): p. 200-208.

150. Sayers, S.P., et al., Effect of ketoprofen on muscle function and sEMG activity after

eccentric exercise. Medicine And Science In Sports And Exercise, 2001. 33(5): p. 702-

710.

151. Hyldahl, R.D., et al., Effects of Ibuprofen Topical Gel on Muscle Soreness. Medicine &

Science in Sports & Exercise, 2010. 42(3): p. 614-621.

152. Krentz, J.R., et al., The effects of ibuprofen on muscle hypertrophy, strength, and

soreness during resistance training. Applied Physiology, Nutrition & Metabolism, 2008.

33(3): p. 470-475.

153. Loram, L.C.M., D.; Fuller, A., Rofecoxib and tramadol do not attenuate delayed-onset

muscle soreness or ischaemic pain in human volunteers. Canadian Journal of Physiology

and Pharmacology, 2005. 83(12): p. 1137-1145.

154. Barlas, P., et al., Managing delayed-onset muscle soreness: lack of effect of selected oral

systemic analgesics. Archives Of Physical Medicine And Rehabilitation, 2000. 81(7): p.

966-972.

155. Grossman, J.M., et al., Effect of ibuprofen use on delayed onset muscle soreness of the

elbow flexors. Journal of Sport Rehabilitation, 1995. 4(4): p. 253-263.

156. Nieman, D.C., et al., Ibuprofen use, endotoxemia, inflammation, and plasma cytokines

during ultramarathon competition. Brain, Behavior, and Immunity, 2006. 20(6): p. 578-

584.

106

157. Semark, A., et al., The effect of a prophylactic dose of flurbiprofen on muscle soreness

and sprinting performance in trained subjects. Journal Of Sports Sciences, 1999. 17(3):

p. 197-203.

158. Stone, M.B., et al., Preliminary comparison of bromelain and Ibuprofen for delayed

onset muscle soreness management. Clinical Journal Of Sport Medicine: Official Journal

Of The Canadian Academy Of Sport Medicine, 2002. 12(6): p. 373-378.

159. Lapointe, B.M., P. Fremont, and C.H. Cote, Influence of nonsteroidal anti-inflammatory

drug treatment duration and time of onset on recovery from exercise-induced muscle

damage in rats. Archives of Physical Medicine & Rehabilitation, 2003. 84(5): p. 651-655.

160. Peterson, J.M., et al., Ibuprofen and acetaminophen: effect on muscle inflammation

after eccentric exercise. Medicine & Science in Sports & Exercise, 2003. 35(6): p. 892-

896.

161. Markworth, J.F., et al., Ibuprofen treatment blunts early translational signaling

responses in human skeletal muscle following resistance exercise. Journal Of Applied

Physiology (Bethesda, Md.: 1985), 2014. 117(1): p. 20-28.

162. Markworth, J.F. and D. Cameron-Smith, Prostaglandin F2α stimulates PI3K/ERK/mTOR

signaling and skeletal myotube hypertrophy. American Journal Of Physiology. Cell

Physiology, 2011. 300(3): p. C671-C682.

163. Rodemann, H.P. and A.L. Goldberg, Arachidonic acid, prostaglandin E2 and F2 alpha

influence rates of protein turnover in skeletal and cardiac muscle. The Journal Of

Biological Chemistry, 1982. 257(4): p. 1632-1638.

164. Shen, W., et al., Inhibited skeletal muscle healing in cyclooxygenase-2 gene-deficient

mice: the role of PGE2 and PGF2alpha. Journal Of Applied Physiology (Bethesda, Md.:

1985), 2006. 101(4): p. 1215-1221.

165. Scott, A., et al., What do we mean by the term "inflammation: a contemporary basic

science update for sports medicine. British Journal of Sports Medicine, 2004. 38(3): p.

372-380.

107

166. Serhan, C.N., Resolvins and protectins: novel lipid mediators in anti-inflammation and

resolution. Scandinavian Journal of Food & Nutrition, 2006. 50: p. 68-78.

167. Serhan, C.N., et al., Resolution of inflammation: state of the art, definitions and terms.

FASEB Journal: Official Publication Of The Federation Of American Societies For

Experimental Biology, 2007. 21(2): p. 325-332.

168. Serhan, C.N., N. Chiang, and T.E. Van Dyke, Resolving inflammation: dual anti-

inflammatory and pro-resolution lipid mediators. Nature Reviews. Immunology, 2008.

8(5): p. 349-361.

169. Serhan, C.N., et al., Novel functional sets of lipid-derived mediators with

antiinflammatory actions generated from omega-3 fatty acids via cyclooxygenase 2-

nonsteroidal antiinflammatory drugs and transcellular processing. The Journal Of

Experimental Medicine, 2000. 192(8): p. 1197-1204.

170. Serhan, C.N., et al., Protectins and maresins: New pro-resolving families of mediators in

acute inflammation and resolution bioactive metabolome. BBA - Molecular & Cell

Biology of Lipids, 2015. 1851(4): p. 397-413.

171. Serhan, C.N., et al., Resolvins: a family of bioactive products of omega-3 fatty acid

transformation circuits initiated by aspirin treatment that counter proinflammation

signals. The Journal Of Experimental Medicine, 2002. 196(8): p. 1025-1037.

172. Serhan, C.N., et al., Design of lipoxin A4 stable analogs that block transmigration and

adhesion of human neutrophils. Biochemistry, 1995. 34(44): p. 14609-14615.

173. Serhan, C.N. and N.A. Petasis, Resolvins and protectins in inflammation resolution.

Chemical Reviews, 2011. 111(10): p. 5922-5943.

174. Serhan, C.N. and J. Savill, Resolution of inflammation: the beginning programs the end.

Nature Immunology, 2005. 6(12): p. 1191-1197.

175. Maddox, J.F. and C.N. Serhan, Lipoxin A4 and B4 are potent stimuli for human monocyte

migration and adhesion: selective inactivation by dehydrogenation and reduction. The

Journal Of Experimental Medicine, 1996. 183(1): p. 137-146.

108

176. Ryan, A. and C. Godson, Lipoxins: regulators of resolution. Current Opinion In

Pharmacology, 2010. 10(2): p. 166-172.

177. Duvall, M.G. and B.D. Levy, DHA- and EPA-derived resolvins, protectins, and maresins in

airway inflammation. European Journal Of Pharmacology, 2015.

178. Hong, S., et al., Novel docosatrienes and 17S-resolvins generated from docosahexaenoic

acid in murine brain, human blood, and glial cells. Autacoids in anti-inflammation. The

Journal Of Biological Chemistry, 2003. 278(17): p. 14677-14687.

179. Bannenberg, G. and C.N. Serhan, Specialized pro-resolving lipid mediators in the

inflammatory response: An update. Biochimica Et Biophysica Acta, 2010. 1801(12): p.

1260-1273.

180. Fadok, V.A., et al., Differential effects of apoptotic versus lysed cells on macrophage

production of cytokines: role of proteases. Journal Of Immunology (Baltimore, Md.:

1950), 2001. 166(11): p. 6847-6854.

181. Godson, C., et al., Cutting edge: lipoxins rapidly stimulate nonphlogistic phagocytosis of

apoptotic neutrophils by monocyte-derived macrophages. Journal Of Immunology

(Baltimore, Md.: 1950), 2000. 164(4): p. 1663-1667.

182. Schwab, J.M., et al., Resolvin E1 and protectin D1 activate inflammation-resolution

programmes. Nature, 2007. 447(7146): p. 869-874.

183. Haslett, C., Granulocyte apoptosis and its role in the resolution and control of lung

inflammation. American Journal Of Respiratory And Critical Care Medicine, 1999. 160(5

Pt 2): p. S5-S11.

184. Serhan, C.N., M. Hamberg, and B. Samuelsson, Lipoxins: novel series of biologically

active compounds formed from arachidonic acid in human leukocytes. Proceedings Of

The National Academy Of Sciences Of The United States Of America, 1984. 81(17): p.

5335-5339.

185. Levy, B.D., et al., Lipid mediator class switching during acute inflammation: signals in

resolution. Nature Immunology, 2001. 2(7): p. 612-619.

109

186. Bannenberg, G.L., et al., Molecular circuits of resolution: formation and actions of

resolvins and protectins. Journal Of Immunology (Baltimore, Md.: 1950), 2005. 174(7):

p. 4345-4355.

187. Gangemi, S., et al., Physical exercise increases urinary excretion of lipoxin A4 and related

compounds. Journal Of Applied Physiology (Bethesda, Md.: 1985), 2003. 94(6): p. 2237-

2240.

188. Baldwin, A.S., Jr., The NF-kappa B and I kappa B proteins: new discoveries and insights.

Annual Review Of Immunology, 1996. 14: p. 649-683.

189. Cai, D., et al., IKKbeta/NF-kappaB activation causes severe muscle wasting in mice. Cell,

2004. 119(2): p. 285-298.

190. Delhalle, S., et al., A beginner's guide to NF-kappaB signaling pathways. Annals Of The

New York Academy Of Sciences, 2004. 1030: p. 1-13.

191. Mourkioti, F. and N. Rosenthal, NF-κB signaling in skeletal muscle: prospects for

intervention in muscle diseases. Journal of Molecular Medicine, 2008. 86(7): p. 747-759.

192. Pahl, H.L., Activators and target genes of Rel/NF-kappaB transcription factors.

Oncogene, 1999. 18(49): p. 6853-6866.

193. Wang, H., et al., NF-kappaB regulation of YY1 inhibits skeletal myogenesis through

transcriptional silencing of myofibrillar genes. Molecular And Cellular Biology, 2007.

27(12): p. 4374-4387.

194. Bakkar, N. and D.C. Guttridge, NF-kappaB signaling: a tale of two pathways in skeletal

myogenesis. Physiological Reviews, 2010. 90(2): p. 495-511.

195. Ghosh, S. and M. Karin, Missing pieces in the NF-kappaB puzzle. Cell, 2002. 109 Suppl:

p. S81-S96.

196. Chen, F.E. and G. Ghosh, Regulation of DNA binding by Rel/NF-kappaB transcription

factors: structural views. Oncogene, 1999. 18(49): p. 6845-6852.

197. Shishodia, S. and B.B. Aggarwal, Nuclear factor-kappaB activation: a question of life or

death. Journal Of Biochemistry And Molecular Biology, 2002. 35(1): p. 28-40.

110

198. Li, H., S. Malhotra, and A. Kumar, Nuclear factor-kappa B signaling in skeletal muscle

atrophy. Journal Of Molecular Medicine (Berlin, Germany), 2008. 86(10): p. 1113-1126.

199. Zandi, E., et al., The IkappaB kinase complex (IKK) contains two kinase subunits,

IKKalpha and IKKbeta, necessary for IkappaB phosphorylation and NF-kappaB

activation. Cell, 1997. 91(2): p. 243-252.

200. Langen, R.C., et al., Inflammatory cytokines inhibit myogenic differentiation through

activation of nuclear factor-kappaB. The FASEB Journal: Official Publication Of The

Federation Of American Societies For Experimental Biology, 2001. 15(7): p. 1169-1180.

201. Layne, M.D., et al., Characterization of the mouse aortic carboxypeptidase-like protein

promoter reveals activity in differentiated and dedifferentiated vascular smooth muscle

cells. Circulation Research, 2002. 90(6): p. 728-736.

202. van de Stolpe, A., et al., 12-O-tetradecanoylphorbol-13-acetate- and tumor necrosis

factor alpha-mediated induction of intercellular adhesion molecule-1 is inhibited by

dexamethasone. Functional analysis of the human intercellular adhesion molecular-1

promoter. The Journal Of Biological Chemistry, 1994. 269(8): p. 6185-6192.

203. Jozci, A.C., et al., Molecular Characteristics of Aged Muscle Reflect an Altered Ability to

Respond to Exercise. International Journal of Sport Nutrition & Exercise Metabolism,

2001. 11(4): p. S9.

204. Ward, C., et al., NF-kappaB activation is a critical regulator of human granulocyte

apoptosis in vitro. The Journal Of Biological Chemistry, 1999. 274(7): p. 4309-4318.

205. Monici, M.C., et al., Activation of nuclear factor-kappaB in inflammatory myopathies

and Duchenne muscular dystrophy. Neurology, 2003. 60(6): p. 993-997.

206. Yang, C.C., et al., Immunolocalization of transcription factor NF-kappaB in inclusion-

body myositis muscle and at normal human neuromuscular junctions. Neuroscience

Letters, 1998. 254(2): p. 77-80.

207. Mourkioti, F., et al., Targeted ablation of IKK2 improves skeletal muscle strength,

maintains mass, and promotes regeneration. The Journal Of Clinical Investigation, 2006.

116(11): p. 2945-2954.

111

208. Fong, C.H.Y., et al., An antiinflammatory role for IKKbeta through the inhibition of

"classical" macrophage activation. The Journal Of Experimental Medicine, 2008. 205(6):

p. 1269-1276.

209. Greten, F.R., et al., NF-kappaB is a negative regulator of IL-1beta secretion as revealed

by genetic and pharmacological inhibition of IKKbeta. Cell, 2007. 130(5): p. 918-931.

210. Lawrence, T., et al., Possible new role for NF-κB in the resolution of inflammation.

Nature Medicine, 2001. 7(12): p. 1291.

211. Lawrence, T. and C. Fong, The resolution of inflammation: anti-inflammatory roles for

NF-kappaB. The International Journal Of Biochemistry & Cell Biology, 2010. 42(4): p.

519-523.

212. Ziegler-Heitbrock, H.W., I. Petersmann, and M. Frankenberger, p50 (NF-kappa B1) is

upregulated in LPS tolerant P388D1 murine macrophages. Immunobiology, 1997. 198(1-

3): p. 73-80.

213. Thaloor, D., et al., Systemic administration of the NF-kappaB inhibitor curcumin

stimulates muscle regeneration after traumatic injury. The American Journal Of

Physiology, 1999. 277(2 Pt 1): p. C320-C329.

214. Guttridge, D.C., et al., NF-kappaB controls cell growth and differentiation through

transcriptional regulation of cyclin D1. Molecular And Cellular Biology, 1999. 19(8): p.

5785-5799.

215. Acharyya, S., et al., Interplay of IKK/NF-kappaB signaling in macrophages and myofibers

promotes muscle degeneration in Duchenne muscular dystrophy. The Journal Of Clinical

Investigation, 2007. 117(4): p. 889-901.

216. Ladner, K.J., M.A. Caligiuri, and D.C. Guttridge, Tumor Necrosis Factor-regulated

Biphasic Activation of NF-κB Is Required for Cytokine-induced Loss of Skeletal Muscle

Gene Products. Journal of Biological Chemistry, 2003. 278(4): p. 2294.

217. Dogra, C., et al., Tumor Necrosis Factor-like Weak Inducer of Apoptosis Inhibits Skeletal

Myogenesis through Sustained Activation of Nuclear Factor-κB and Degradation of

MyoD Protein. Journal of Biological Chemistry, 2006. 281(15): p. 10327-10336.

112

218. Szalay, K., Z. Rázga, and E. Duda, TNF inhibits myogenesis and downregulates the

expression of myogenic regulatory factors myoD and myogenin. European Journal Of

Cell Biology, 1997. 74(4): p. 391-398.

219. Hirasaka, K., et al., Clinorotation prevents differentiation of rat myoblastic L6 cells in

association with reduced NF-kappa B signaling. Biochimica Et Biophysica Acta, 2005.

1743(1-2): p. 130-140.

220. Kim, S.S., et al., Inhibitors of the proteasome block the myogenic differentiation of rat L6

myoblasts. FEBS Letters, 1998. 433(1-2): p. 47-50.

221. Olashaw, N.E., et al., Induction of NF-kappa B-like activity by platelet-derived growth

factor in mouse fibroblasts. Molecular Biology Of The Cell, 1992. 3(10): p. 1131-1139.

222. Canicio, J., et al., Nuclear factor kappa B-inducing kinase and Ikappa B kinase-alpha

signal skeletal muscle cell differentiation. The Journal Of Biological Chemistry, 2001.

276(23): p. 20228-20233.

223. Frost, R.A., G.J. Nystrom, and C.H. Lang, Stimulation of insulin-like growth factor binding

protein-1 synthesis by interleukin-1beta: requirement of the mitogen-activated protein

kinase pathway. Endocrinology, 2000. 141(9): p. 3156-3164.

224. Kaliman, P., et al., Insulin-like growth factor-II, phosphatidylinositol 3-kinase, nuclear

factor-kappaB and inducible nitric-oxide synthase define a common myogenic signaling

pathway. The Journal Of Biological Chemistry, 1999. 274(25): p. 17437-17444.

225. Baeza-Raja, B. and P. Muñoz-Cánoves, p38 MAPK-induced nuclear factor-kappaB

activity is required for skeletal muscle differentiation: role of interleukin-6. Molecular

Biology Of The Cell, 2004. 15(4): p. 2013-2026.

226. De Alvaro, C., et al., Nuclear exclusion of forkhead box O and Elk1 and activation of

nuclear factor-kappaB are required for C2C12-RasV12C40 myoblast differentiation.

Endocrinology, 2008. 149(2): p. 793-801.

227. Madrid, L.V., et al., Akt stimulates the transactivation potential of the RelA/p65 Subunit

of NF-kappa B through utilization of the Ikappa B kinase and activation of the mitogen-

113

activated protein kinase p38. The Journal Of Biological Chemistry, 2001. 276(22): p.

18934-18940.

228. Chen, S.-E., B. Jin, and Y.-P. Li, TNF-alpha regulates myogenesis and muscle

regeneration by activating p38 MAPK. American Journal Of Physiology. Cell Physiology,

2007. 292(5): p. C1660-C1671.

229. Collins, R.A. and M.D. Grounds, The role of tumor necrosis factor-alpha (TNF-alpha) in

skeletal muscle regeneration. Studies in TNF-alpha(-/-) and TNF-alpha(-/-)/LT-alpha(-/-)

mice. The Journal Of Histochemistry And Cytochemistry: Official Journal Of The

Histochemistry Society, 2001. 49(8): p. 989-1001.

230. Chen, S.-E., et al., Role of TNF-{alpha} signaling in regeneration of cardiotoxin-injured

muscle. American Journal Of Physiology. Cell Physiology, 2005. 289(5): p. C1179-C1187.

231. Purcell, N.H., et al., Activation of NF-kappa B is required for hypertrophic growth of

primary rat neonatal ventricular cardiomyocytes. Proceedings Of The National Academy

Of Sciences Of The United States Of America, 2001. 98(12): p. 6668-6673.

232. García-López, D., et al., Effects of eccentric exercise on NF-kappaB activation in blood

mononuclear cells. Medicine And Science In Sports And Exercise, 2007. 39(4): p. 653-

664.

233. Ho, R.C., et al., Regulation of IkappaB kinase and NF-kappaB in contracting adult rat

skeletal muscle. American Journal Of Physiology. Cell Physiology, 2005. 289(4): p. C794-

C801.

234. Ji, L.L., et al., Acute exercise activates nuclear factor (NF)-kappaB signaling pathway in

rat skeletal muscle. The FASEB Journal: Official Publication Of The Federation Of

American Societies For Experimental Biology, 2004. 18(13): p. 1499-1506.

235. Jiménez-Jiménez, R., et al., Eccentric training impairs NF-kappaB activation and over-

expression of inflammation-related genes induced by acute eccentric exercise in the

elderly. Mechanisms Of Ageing And Development, 2008. 129(6): p. 313-321.

114

236. Tantiwong, P., et al., NF-κB activity in muscle from obese and type 2 diabetic subjects

under basal and exercise-stimulated conditions. American Journal Of Physiology.

Endocrinology And Metabolism, 2010. 299(5): p. E794-E801.

237. Lille, S.T., et al., Inhibition of the initial wave of NF-kappaB activity in rat muscle reduces

ischemia/reperfusion injury. Muscle & Nerve, 2001. 24(4): p. 534-541.

238. Durham, W.J., et al., Fatiguing exercise reduces DNA binding activity of NF-κB in skeletal

muscle nuclei. Journal of Applied Physiology, 2004. 97(5): p. 1740-1745.

239. Vella, L., et al., Resistance exercise increases NF-κB activity in human skeletal muscle.

American Journal Of Physiology. Regulatory, Integrative And Comparative Physiology,

2012. 302(6): p. R667-R673.

240. Faulkner, J.A., S.V. Brooks, and J.A. Opiteck, Injury to skeletal muscle fibers during

contractions; conditions of occurrence and prevention. Physical Therapy, 1993. 73(12):

p. 911-921.

241. Baldwin, A.C., S.W. Stevenson, and G.A. Dudley, Nonsteroidal anti-inflammatory

therapy after eccentric exercise in healthy older individuals. Journals of Gerontology

Series A: Biological Sciences & Medical Sciences, 2001. 56(8): p. M510-m513.

242. Hersh, E.V., P.A. Moore, and G.L. Ross, Over-the-counter analgesics and antipyretics: a

critical assessment. Clinical Therapeutics, 2000. 22(5): p. 500-548.

243. Mikkelsen, U.R., et al., Local NSAID infusion does not affect protein synthesis and gene

expression in human muscle after eccentric exercise. Scandinavian Journal of Medicine

& Science in Sports, 2011. 21(5): p. 630-644.

244. Whisenant, M.J., et al., Validation of submaximal prediction equations for the 1

repetition maximum bench press test on a group of collegiate football players. Journal

Of Strength And Conditioning Research / National Strength & Conditioning Association,

2003. 17(2): p. 221-227.

245. Buford, T.W., M.B. Cooke, and D.S. Willoughby, Resistance exercise-induced changes of

inflammatory gene expression within human skeletal muscle. European Journal Of

Applied Physiology, 2009. 107(4): p. 463-471.

115

246. Vella, L., et al., Ibuprofen supplementation and its effects on NF-κB activation in skeletal

muscle following resistance exercise. Physiological Reports, 2014. 2(10).

247. MacNeil, L.G., et al., 17β-estradiol attenuates exercise-induced neutrophil infiltration in

men. American Journal Of Physiology. Regulatory, Integrative And Comparative

Physiology, 2011. 300(6): p. R1443-R1451.

248. Lapointe, B.M., P. Frémont, and C.H. Côté, Adaptation to lengthening contractions is

independent of voluntary muscle recruitment but relies on inflammation. American

Journal Of Physiology. Regulatory, Integrative And Comparative Physiology, 2002.

282(1): p. R323-R329.

249. Lapointe, B.M., J. Frenette, and C.H. Côté, Lengthening contraction-induced

inflammation is linked to secondary damage but devoid of neutrophil invasion. Journal

Of Applied Physiology (Bethesda, Md.: 1985), 2002. 92(5): p. 1995-2004.

250. Nosaka, K., P. Sacco, and K. Mawatari, Effects of Amino Acid Supplementation on Muscle

Soreness and Damage. International Journal of Sport Nutrition & Exercise Metabolism,

2006. 16(6): p. 620-635.

251. Connolly, D.A.J., S.P. Sayers, and M.P. McHugh, Treatment and prevention of delayed

onset muscle soreness. Journal of Strength & Conditioning Research (Allen Press

Publishing Services Inc.), 2003. 17(1): p. 197-208.

252. Malm, C., et al., Leukocytes, cytokines, growth factors and hormones in human skeletal

muscle and blood after uphill or downhill running. The Journal Of Physiology, 2004.

556(Pt 3): p. 983-1000.

253. Gibson, W., et al., Increased pain from muscle fascia following eccentric exercise: animal

and human findings. Experimental Brain Research, 2009. 194(2): p. 299-308.

254. Raastad, T., et al., Changes in Calpain Activity, Muscle Structure, and Function after

Eccentric Exercise. Medicine & Science in Sports & Exercise, 2010. 42(1): p. 86-95.

255. Roberts, L.A., et al., Post-exercise cold water immersion attenuates acute anabolic

signalling and long-term adaptations in muscle to strength training. The Journal Of

Physiology, 2015. 593(18): p. 4285-4301.

116

256. Farnfield, M.M., et al., Whey protein ingestion activates mTOR-dependent signalling

after resistance exercise in young men: a double-blinded randomized controlled trial.

Nutrients, 2009. 1(2): p. 263-275.

257. Trenerry, M.K., et al., STAT3 signaling is activated in human skeletal muscle following

acute resistance exercise. J Appl Physiol, 2007. 102(4): p. 1483-9.

258. Markworth, J.F., et al., Human inflammatory and resolving lipid mediator responses to

resistance exercise and ibuprofen treatment. Am J Physiol Regul Integr Comp Physiol,

2013. 305(11): p. R1281-96.

259. Maddipati, K.R., et al., Eicosanomic profiling reveals dominance of the epoxygenase

pathway in human amniotic fluid at term in spontaneous labor. FASEB J, 2014. 28(11):

p. 4835-46.

260. Maddipati, K.R., et al., Clinical chorioamnionitis at term: the amniotic fluid fatty acyl

lipidome. Journal of Lipid Research, 2016. 57(10): p. 1906-1916.

261. Haeggström, J.Z. and C.D. Funk, Lipoxygenase and Leukotriene Pathways: Biochemistry,

Biology, and Roles in Disease. Chemical Reviews, 2011. 111(10): p. 5866-5898.

262. Dobrian, A.D., et al., Functional and pathological roles of the 12- and 15-lipoxygenases.

Progress In Lipid Research, 2011. 50(1): p. 115-131.

263. Yeung, J. and M. Holinstat, 12-lipoxygenase: a potential target for novel anti-platelet

therapeutics. Cardiovascular & Hematological Agents In Medicinal Chemistry, 2011.

9(3): p. 154-164.

264. Yin, B., et al., Arachidonate 12-lipoxygenase may serve as a potential marker and

therapeutic target for prostate cancer stem cells. International Journal Of Oncology,

2011. 38(4): p. 1041-1046.

265. Wuest, S.J.A., et al., Expression and regulation of 12/15-lipoxygenases in human

primary macrophages. Atherosclerosis, 2012. 225(1): p. 121-127.

266. Markworth, J.F. and D. Cameron-Smith, Prostagiandin F2α stimulates PI3K/ERK/mTOR

signaling and skeletal myotube hypertrophy. American Journal of Physiology: Cell

Physiology, 2011. 69(3): p. C671-C682.

117

267. Laustiola, K., et al., Exercise-induced increase in plasma arachidonic acid and

thromboxane B2 in healthy men: effect of beta-adrenergic blockade. Journal Of

Cardiovascular Pharmacology, 1984. 6(3): p. 449-454.

268. Hilberg, T., et al., Transcription in response to physical stress--clues to the molecular

mechanisms of exercise-induced asthma. FASEB Journal: Official Publication Of The

Federation Of American Societies For Experimental Biology, 2005. 19(11): p. 1492-1494.

269. Massoumi, R. and A. Sjölander, The role of leukotriene receptor signaling in

inflammation and cancer. Thescientificworldjournal, 2007. 7: p. 1413-1421.

270. Rouzer, C.A., et al., Cloning and expression of human leukocyte 5-lipoxygenase.

Advances In Prostaglandin, Thromboxane, And Leukotriene Research, 1989. 19: p. 474-

477.

271. Reynaud, D. and C.R. Pace-Asciak, 12-HETE and 12-HPETE potently stimulate

intracellular release of calcium in intact human neutrophils. Prostaglandins,

Leukotrienes, And Essential Fatty Acids, 1997. 56(1): p. 9-12.

272. Romano, M., et al., Lipoxin synthase activity of human platelet 12-lipoxygenase. The

Biochemical Journal, 1993. 296 ( Pt 1): p. 127-133.

273. Serhan, C.N. and K.A. Sheppard, Lipoxin formation during human neutrophil-platelet

interactions. Evidence for the transformation of leukotriene A4 by platelet 12-

lipoxygenase in vitro. The Journal Of Clinical Investigation, 1990. 85(3): p. 772-780.

274. Serhan, C.N., On the relationship between leukotriene and lipoxin production by human

neutrophils: evidence for differential metabolism of 15-HETE and 5-HETE. Biochimica Et

Biophysica Acta, 1989. 1004(2): p. 158-168.

275. Spector, A.A., et al., Epoxyeicosatrienoic acids (EETs): metabolism and biochemical

function. Progress In Lipid Research, 2004. 43(1): p. 55-90.

276. Konkel, A. and W.-H. Schunck, Role of cytochrome P450 enzymes in the bioactivation of

polyunsaturated fatty acids. Biochimica Et Biophysica Acta, 2011. 1814(1): p. 210-222.

118

277. Nieman, D.C., et al., Influence of pistachios on performance and exercise-induced

inflammation, oxidative stress, immune dysfunction, and metabolite shifts in cyclists: a

randomized, crossover trial. Plos One, 2014. 9(11): p. e113725-e113725.

278. Thompson, D.A. and B.D. Hammock, Dihydroxyoctadecamonoenoate esters inhibit the

neutrophil respiratory burst. Journal of Biosciences, 2007. 32(2): p. 279-291.

279. Paulsen, G., et al., Leucocytes, cytokines and satellite cells: what role do they play in

muscle damage and regeneration following eccentric exercise? Exercise Immunology

Review, 2012. 18: p. 42-97.

280. Trappe, T.A., et al., Effect of ibuprofen and acetaminophen on postexercise muscle

protein synthesis. American Journal of Physiology: Endocrinology & Metabolism, 2002.

45(3): p. E551.

281. Bonizzi, G. and M. Karin, The two NF-kappaB activation pathways and their role in

innate and adaptive immunity. Trends In Immunology, 2004. 25(6): p. 280-288.

282. Senftleben, U., et al., Activation by IKKalpha of a second, evolutionary conserved, NF-

kappa B signaling pathway. Science (New York, N.Y.), 2001. 293(5534): p. 1495-1499.

283. Lawrence, T., et al., Possible new role for NF-kappaB in the resolution of inflammation.

Nature Medicine, 2001. 7(12): p. 1291-1297.

284. Bohuslav, J., et al., Regulation of an essential innate immune response by the p50

subunit of NF-kappaB. The Journal Of Clinical Investigation, 1998. 102(9): p. 1645-1652.

285. Ishikawa, H., et al., Chronic inflammation and susceptibility to bacterial infections in

mice lacking the polypeptide (p)105 precursor (NF-kappaB1) but expressing p50. The

Journal Of Experimental Medicine, 1998. 187(7): p. 985-996.

286. Thomas, B., et al., Critical role of C/EBPdelta and C/EBPbeta factors in the stimulation of

the cyclooxygenase-2 gene transcription by interleukin-1beta in articular chondrocytes.

European Journal Of Biochemistry / FEBS, 2000. 267(23): p. 6798-6809.

287. Wadleigh, D.J., et al., Transcriptional activation of the cyclooxygenase-2 gene in

endotoxin-treated RAW 264.7 macrophages. The Journal Of Biological Chemistry, 2000.

275(9): p. 6259-6266.

119

288. Hollander, J., et al., Superoxide dismutase gene expression is activated by a single bout

of exercise in rat skeletal muscle. Pflügers Archiv: European Journal Of Physiology, 2001.

442(3): p. 426-434.

289. Kramer, H.F. and L.J. Goodyear, Exercise, MAPK, and NF-kappaB signaling in skeletal

muscle. Journal Of Applied Physiology (Bethesda, Md.: 1985), 2007. 103(1): p. 388-395.

290. Spangenburg, E.E., et al., Exercise increases SOCS-3 expression in rat skeletal muscle:

potential relationship to IL-6 expression. The Journal Of Physiology, 2006. 572(Pt 3): p.

839-848.

291. Mayhew, J.L., et al., Muscular endurance repetitions to predict bench press strength in

men of different training levels. The Journal Of Sports Medicine And Physical Fitness,

1995. 35(2): p. 108-113.

292. Mead, R., The design of experiments: Statistical principles for practical applications. The

design of experiments: Statistical principles for practical applications. 1988, New York,

NY US: Cambridge University Press.

293. Saville, D.J., Multiple comparison procedures: The practical solution. American

Statistician, 1990. 44(2): p. 174.

294. Hyldahl, R.D., et al., Activation of nuclear factor-κB following muscle eccentric

contractions in humans is localized primarily to skeletal muscle-residing pericytes.

FASEB Journal: Official Publication Of The Federation Of American Societies For

Experimental Biology, 2011. 25(9): p. 2956-2966.

295. Xin, L., et al., A contralateral repeated bout effect attenuates induction of NF-κB DNA-

binding following eccentric exercise. Journal Of Applied Physiology (Bethesda, Md.:

1985), 2013.

296. Lawrence, T., et al., IKKalpha limits macrophage NF-kappaB activation and contributes

to the resolution of inflammation. Nature, 2005. 434(7037): p. 1138-1143.

297. Poligone, B. and A.S. Baldwin, Positive and negative regulation of NF-kappaB by COX-2:

roles of different prostaglandins. The Journal Of Biological Chemistry, 2001. 276(42): p.

38658-38664.

120

298. Rossi, A., et al., Anti-inflammatory cyclopentenone prostaglandins are direct inhibitors

of IkappaB kinase. Nature, 2000. 403(6765): p. 103-108.

299. Straus, D.S., et al., 15-deoxy-delta 12,14-prostaglandin J2 inhibits multiple steps in the

NF-kappa B signaling pathway. Proceedings Of The National Academy Of Sciences Of

The United States Of America, 2000. 97(9): p. 4844-4849.

300. Castrillo, A., et al., Inhibition of IkappaB kinase and IkappaB phosphorylation by 15-

deoxy-Delta(12,14)-prostaglandin J(2) in activated murine macrophages. Molecular And

Cellular Biology, 2000. 20(5): p. 1692-1698.

301. Lawrence, T., D.A. Willoughby, and D.W. Gilroy, Anti-inflammatory lipid mediators and

insights into the resolution of inflammation. Nature Reviews. Immunology, 2002. 2(10):

p. 787-795.

302. Brickson, S., et al., M1/70 attenuates blood-borne neutrophil oxidants, activation, and

myofiber damage following stretch injury. Journal Of Applied Physiology (Bethesda,

Md.: 1985), 2003. 95(3): p. 969-976.

303. Tidball, J.G., Interactions between muscle and the immune system during modified

musculoskeletal loading. Clinical Orthopedics, 2002. 403(suppl): p. S100-S109.

304. Veliça, P. and C.M. Bunce, Prostaglandins in muscle regeneration. Journal Of Muscle

Research And Cell Motility, 2008. 29(6-8): p. 163-167.

305. Arita, M., et al., Stereochemical assignment, antiinflammatory properties, and receptor

for the omega-3 lipid mediator resolvin E1. Journal of Experimental Medicine, 2005.

201(5): p. 713-722.

306. Bandeira-Melo, C., et al., Cyclooxygenase-2-derived prostaglandin E2 and lipoxin A4

accelerate resolution of allergic edema in Angiostrongylus costaricensis-infected rats:

relationship with concurrent eosinophilia. Journal of Immunology (Baltimore), 2000.

164(2): p. 1029-1036.

307. Markworth, J.F., et al., Divergent shifts in lipid mediator profile following

supplementation with n-3 docosapentaenoic acid and eicosapentaenoic acid. FASEB

121

Journal: Official Publication Of The Federation Of American Societies For Experimental

Biology, 2016.

308. Miokovic, T., et al., Heterogeneous atrophy occurs within individual lower limb muscles

during 60 days of bed rest. Journal Of Applied Physiology (Bethesda, Md.: 1985), 2012.

113(10): p. 1545-1559.

309. Lexell, J., K. Henriksson-Larsén, and M. Sjöström, Distribution of different fibre types in

human skeletal muscles. 2. A study of cross-sections of whole m. vastus lateralis. Acta

Physiologica Scandinavica, 1983. 117(1): p. 115-122.

122

Appendix A:

ORIGINAL RESEARCHpublished: 29 March 2016

doi: 10.3389/fphys.2016.00086

Frontiers in Physiology | www.frontiersin.org 1 March 2016 | Volume 7 | Article 86

Edited by:

Vincent Pialoux,

University Lyon 1, France

Reviewed by:

Gianluca Vernillo,

University of Milan, Italy

Scott Forbes,

Okanagan College, Canada

*Correspondence:

Luke Vella

[email protected]

Specialty section:

This article was submitted to

Exercise Physiology,

a section of the journal

Frontiers in Physiology

Received: 12 December 2015

Accepted: 22 February 2016

Published: 29 March 2016

Citation:

Vella L, Markworth JF, Paulsen G,

Raastad T, Peake JM, Snow RJ,

Cameron-Smith D and Russell AP

(2016) Ibuprofen Ingestion Does Not

Affect Markers of Post-exercise

Muscle Inflammation.

Front. Physiol. 7:86.

doi: 10.3389/fphys.2016.00086

Ibuprofen Ingestion Does Not AffectMarkers of Post-exercise MuscleInflammationLuke Vella 1*, James F. Markworth 2, Gøran Paulsen 3, Truls Raastad 3, Jonathan M. Peake 4,

Rod J. Snow 1, David Cameron-Smith 2 and Aaron P. Russell 1

1Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Science, Deakin University, Burwood,

VIC, Australia, 2 Liggins Institute, University of Auckland, Auckland, New Zealand, 3Department of Physical Performance,

Norwegian School of Sport Science, Oslo, Norway, 4 School of Biomedical Sciences and Institute of Health and Biomedical

Innovation, Queensland University of Technology, Brisbane, QLD, Australia

Purpose: We investigated if oral ingestion of ibuprofen influenced leucocyte recruitment

and infiltration following an acute bout of traditional resistance exercise.

Methods: Sixteen male subjects were divided into two groups that received the

maximum over-the-counter dose of ibuprofen (1200mg d−1) or a similarly administered

placebo following lower body resistance exercise. Muscle biopsies were taken from

m.vastus lateralis and blood serum samples were obtained before and immediately after

exercise, and at 3 and 24 h after exercise. Muscle cross-sections were stained with

antibodies against neutrophils (CD66b and MPO) and macrophages (CD68). Muscle

damage was assessed via creatine kinase and myoglobin in blood serum samples, and

muscle soreness was rated on a ten-point pain scale.

Results: The resistance exercise protocol stimulated a significant increase in the number

of CD66b+ and MPO+ cells when measured 3 h post exercise. Serum creatine kinase,

myoglobin and subjective muscle soreness all increased post-exercise. Muscle leucocyte

infiltration, creatine kinase, myoglobin and subjective muscle soreness were unaffected

by ibuprofen treatment when compared to placebo. There was also no association

between increases in inflammatory leucocytes and any other marker of cellular muscle

damage.

Conclusion: Ibuprofen administration had no effect on the accumulation of neutrophils,

markers of muscle damage or muscle soreness during the first 24 h of post-exercise

muscle recovery.

Keywords: exercise recovery, NSAID treatment, inflammation, resistance exercise, leucocyte

INTRODUCTION

Unaccustomed resistance exercise often results in tissue damage and inflammation, leadingto delayed onset muscle soreness (DOMS) and a consequent reduction in force production(Faulkner et al., 1993; Tidball, 1995). Animal models and in vitro studies have identified that localand systemic inflammation exerts a regulatory influence during the different phases of musclerecovery, including myofibrillar disruption, cellular necrosis, satellite cell activation, maturationand subsequent regeneration, and adaptation (Fridén et al., 1981; Armstrong et al., 1991). Thus,

Vella et al. Ibuprofen and Post-exercise Inflammation

post-exercise inflammation is intimately necessary and a keyfeature of the normal process of tissue regeneration andadaptation following acute muscle damage. However, excessiveinflammation is considered a potential cause of prolongedpost-exercise muscle soreness and may have a negative effecton muscle recovery (Armstrong et al., 1991; Smith, 1991);consequently strategies to reduce or counteract inflammation arecommonly implemented to aid in improving muscle recoveryafter exercise (Urso, 2013).

Non-steroidal anti-inflammatory drugs (NSAIDs) arecommonly used as a treatment strategy in exercise andsports medicine to assist with recovery from exercise-inducedinflammation, particularly following soft-tissue injury. NSAIDsinhibit the cyclooxygenase (COX-1 and 2) enzymes andconsequently the formation of prostanoids (prostaglandins,prostacyclins, and thromboxanes) that play a diverse role inacute inflammation (Markworth et al., 2013). Prostanoidsstimulate an acute inflammatory process by controlling localblood flow, vascular permeability, leucocyte infiltration, andtriggering sensations of pain (Markworth et al., 2013; Urso,2013). In animal models of acute muscle damage, treatmentwith NSAIDs blunts the infiltration of leucocytes into muscletissue (Lapointe et al., 2003; Bondesen et al., 2004) and causesa reduction in creatine kinase (CK) (Mishra et al., 1995).Consequently, NSAIDs can also inhibit myofiber regeneration,satellite cell proliferation and differentiation, and overload-induced muscle hypertrophy (Mishra et al., 1995; Bondesenet al., 2004, 2006). These findings provide preliminary evidencethat NSAIDs compromise the physiological link betweenprocesses of acute muscle damage, inflammation and cellularregeneration.

Research into the effects of NSAIDs on exercise-inducedmuscle damage and inflammation has produced equivocalfindings. In exercise models, NSAID administration has beenshown to attenuate post-exercise DOMS in some (Baldwin et al.,2001; Tokmakidis et al., 2003; Paulsen et al., 2010), but notall studies (Trappe et al., 2002; Krentz et al., 2008; Mikkelsenet al., 2009; Hyldahl et al., 2010). While a precise cellularmechanism for an analgesic effect of NSAIDs remains unclear,it has been largely ascribed to their effect on prostaglandinsynthesis and the capacity of NSAIDs to interfere with aspectsof inflammatory cell function (Hersh et al., 2000; Petersonet al., 2003). Previous research has demonstrated that oralconsumption of both ibuprofen, a non-selective NSAID andacetaminophen, an analgesic also known as paracetamol, had noeffect on macrophage infiltration at 24 h following an eccentricexercise protocol (Peterson et al., 2003). Similarly treatment withnaproxen, another non-selective NSAID, had no effect on theinfiltration of leucocyte common antigen positive cells followinga unilateral, isotonic resistance exercise protocol (Bourgeois et al.,1999). Interestingly, Paulsen et al. (2010) suggested a bluntingeffect of the COX-2 specific celecoxib following maximumeccentric muscle contractions (Paulsen et al., 2010). This researchidentified a tendency for higher monocyte/macrophage numbersin subjects within the placebo group, and those subjects who wereidentified as “high-responders” to the exercise protocol based onthe number of inflammatory leucocytes (Paulsen et al., 2010).

Although this is not a particularly robust finding, it has led to thehypothesis that NSAIDs may influence leucocyte infiltration inskeletal muscle when a sufficiently strong and early inflammatoryreaction is present (Paulsen et al., 2010). This hypothesis wouldsuggest that the intensity of the exercise stimulus and theconsequent muscle-damage response would be largely influentialin determining the effect of a pharmacologically based anti-inflammatory intervention.

Conflicting findings also exist with regard to the effectof NSAIDs on the regenerative capacity of skeletal musclefollowing exercise-induced muscle damage. The non-selectiveCOX inhibitor ibuprofen blunted skeletal muscle proteinsynthesis (Trappe et al., 2002) while local intramuscularinfusion of indomethacin (Mikkelsen et al., 2011) and theoral administration of the COX-2 selective NSAID celecoxib(Burd et al., 2010; Paulsen et al., 2010) had no such effect.Similarly, treatment with indomethacin inhibited post-exercisesatellite cell proliferation (Mikkelsen et al., 2009). Recent workfrom our group demonstrated that treatment with ibuprofeninhibited early translational signaling responses involved in post-exercise muscle hypertrophy (Markworth et al., 2014). A clearmechanistic pathway for NSAIDs to influence the physiologicallink between post-exercise inflammation and skeletal muscleregeneration remains elusive.

These discrepancies in the research to date are likely dueto differences in the exercise protocol (concentric vs. eccentricmuscle contractions), the training status of subjects, the timingof muscle biopsies, and the type of NSAID, the dosageadministered, and method of administration. Further research isrequired to determine whether NSAID administration affects theinfiltration of leucocyte populations following exercise-inducedmuscle damage and how this influences post-exercise adaptivepathways. The aim of the present study was to investigate iforal administration of the NSAID ibuprofen influenced skeletalmuscle leucocyte infiltration during the first 24 h after muscle-damaging resistance exercises. We also aimed to explore howany changes in leucocyte infiltration were related to markersof muscle damage, including circulating muscle proteins CKand myoglobin, and subjective markers of muscle soreness.We hypothesized that the ingestion of ibuprofen followingacute resistance exercise would not influence the infiltration ofleucocytes, however it would attenuate sensations of DOMS.

MATERIALS AND METHODS

ParticipantsAs described previously, 16 healthy male subjects were recruitedto participate in the study (Table 1; Markworth et al., 2013). Itis important to note that the purpose of the original study byMarkworth et al. (2013) was to profile the human eicosanoidresponse to acute exercise in blood plasma samples, as opposedto the aim of the present study which was to determine theeffect of oral ingestion of ibuprofen on leucocyte recruitmentand infiltration following an acute bout of traditional resistanceexercise.While the subjects and experimental design are the samein both studies the aims and dependent variables reported aredifferent. Exclusion criteria included participation in a lower

Frontiers in Physiology | www.frontiersin.org 2 March 2016 | Volume 7 | Article 86

Vella et al. Ibuprofen and Post-exercise Inflammation

TABLE 1 | Subject characteristics and strength testing data.

Characteristics Strength (1 RM)

Age (y) Height (m) Body mass (kg) BMI Squat (kg) Leg press (kg) Leg extension (kg)

PLA 23.9 ± 1.3 1.89 ± 0.1 86.9 ± 4.5 24.5 ± 1.2 94.9 ± 5.4 237 ± 17 236 ± 18

IBU 23 ± 0.5 1.89 ± 0.1 89.1 ± 4.4 24.8 ± 0.8 91.9 ± 6.0 240 ± 15 196 ± 22

Values are mean values ± SEM. No significant differences were observed between the two groups. PLA, placebo; IBU, Ibuprofen; BMI, body mass index.

body resistance exercise program within the last 6 months toensure a muscle damage response from the exercise stimulus,and/or previous chronic treatments with anti-inflammatorymedication. Participants also completed a medical screening toidentify any potential risk factors for them to perform strenuousphysical activity.

Ethics ApprovalAll procedures involved in this study were approved bythe Deakin University Human Research Ethics Committee(DUHREC 2010-019) and muscle sampling procedures wereperformed in accordance with the Helsinki declaration. Eachparticipant was provided with written and oral details of thenature and requirements of the study and provided writtenconsent to participate.

FamiliarizationEach participant completed a familiarization and strength testingsession at least 7 days prior to completing the exercise protocol.Details of the familiarization session have been describedpreviously (Markworth et al., 2013). Briefly, subjects performedrepetition maximum testing for the Smith machine-assistedsquat, the leg press and the leg extension to determine theirexperimental exercise load [80% of a 1 repetition maximum (1RM)]. The maximum weight the subject could lift for 5–8 repswas determined, and these data were entered into the Brzyckiequation to predict 1 RM (Whisenant et al., 2003). Subjects wereasked to abstain from any further activity until the completion ofthe trial.

Experimental ProceduresThe participants reported to the laboratory on the morningof the trial in an overnight fasted state. They were asked toabstain from caffeine, tobacco and alcohol for the 24 h precedingthe trial. Participants rested in a supine position for 30min,following which the first muscle biopsy sample was taken.Each participant then completed a 10min warm-up protocolcomprised of 5min of low intensity cycling on a stationarybike, and one low-intensity set of each exercise at a weight ofeach subject’s own choice within the range of 30–50% 1 RM.The resistance exercise session consisted of three sets of 8–10repetitions performed on a Smith machine assisted squat, a 45◦

leg press and a leg extension at 80% of a predicted 1 RM. Theexercises were performed as a circuit with 1min rest permittedbetween exercises and 3min rest between sets. This protocolhas been used previously and has been a sufficient stimulus toactivate inflammatory signaling pathways (Vella et al., 2012) and

was implemented to replicate a commonly used exercise routine.After exercise, the subjects rested while subsequentmuscle biopsysamples were collected. After the 3 h biopsy, participants wereprovided a standardized meal and were allowed to go home.The following morning, participants reported to the laboratoryin an over-night fasted state for a 24 h muscle biopsy and bloodsample.

Standardized MealsStandardized meals were provided to participants on the nightbefore the trial, (carbohydrate 57%, fat 22%, protein 21%),immediately following the 3 h muscle biopsy (carbohydrate 71%,fat 13%, protein 18%), and in the evening (carbohydrate 64%, fat27%, protein 18%) on the day of the exercise trial. Participantswere permitted to drinkwater ad libitum andwere asked to reportif they could not finish their allocated meals. This nutrition planwas included to ensure that each participant received the samerelative percentage of macro- and micro-nutrients.

NSAID AdministrationPrior to exercise, participants were randomly assigned ina double-blind method, to consume either the maximumrecommended dose of ibuprofen (IBU, n = 8) or a placebocontrol (gelatin capsules identical in appearance containingpowdered sugar in place of ibuprofen) (PLA, n = 8). The IBUgroup consumed three doses of 400mg of ibuprofen throughoutthe trial day. The first dose was administered immediately priorto the first muscle biopsy sample. Participants were instructedto consume the following two doses at 6 and 12 h followingthe exercise protocol. Follow-up phone calls from the researchteam ensured compliance. To ensure this dosing structure wasappropriate to maintain biologically active levels of ibuprofen,ibuprofen concentration was measured in blood serum samplesand these data are reported elsewhere (Markworth et al.,2013).

Sample CollectionVenous blood samples were drawn prior to exercise, within 5minpost exercise (here-after referred to as 0 h post-exercise), and at 1,2, 3, and 24 h post-exercise. Blood samples were drawn throughan indwelling catheter into VACUETTE serum tubes (Greiner,Bio-One, Stonehouse UK). Whole blood was allowed to clot atroom temperature. Samples were then centrifuged at 1000 × gfor 10min. The serum layer was collected and stored at −80◦Cfor subsequent analysis.

Muscle biopsy samples were obtained from the vastuslateralis muscle under local anesthesia (Xylocaine 1%) using

Frontiers in Physiology | www.frontiersin.org 3 March 2016 | Volume 7 | Article 86

Vella et al. Ibuprofen and Post-exercise Inflammation

a percutaneous needle biopsy technique modified to includesuction (Buford et al., 2009). To minimize interference fromthe biopsy procedure, samples prior to exercise and at 24 h postexercise were taken from the same leg, while samples taken at0 and 3 h post-exercise were taken from the contralateral leg.Samples obtained within the same leg were taken at least 5 cmfrom the previous incision site. We have previously reportedthat this technique is effective for minimizing the risk of anyinflammation arising from the biopsy procedure confoundingexercise-induced inflammation (Vella et al., 2012). Tissue-Tekimmersed tissue was rapidly frozen in isopentane cooled liquidnitrogen before storage at−80◦C.

ImmunohistochemistryCross sections (8µm) of muscle tissue were cut using amicrotome at −20◦C (CM3050, Leica, Germany) and mountedon microscope slides (Superfrost Plus, Thermo Scientific,MA, USA), air-dried and stored at −80◦C. Each subject’smuscle sections from all time points were mounted on thesame microscope slide. Serial muscle cross-sections werestained with three different leucocyte antibodies, MPO (#0398,DakoCytomation, Glostrup, Denmark; dilution 1:2000), CD66b(#CLB-B13.9, Sanquin Reagents, Amsterdam, The Netherlands;dilution 1:500), and CD68 (#EBM-11, DakoCytomation,Glostrup, Denmark; dilution 1:300). Furthermore, the sectionswere also stained with dystrophin antibody (ab15277, Abcam,Cambridge, UK) for visualizing borders of muscle fibers.

Sections were fixed in 4% paraformaldehyde solution in astaining jar for 5min at room temperature and rinsed twicefor 10min in phosphate buffered saline (PBS) containing 0.5%Tween 20 (Sigma-Aldrich). The microscope slides were movedinto a humidified chamber and non-specific binding sites wereblocked with 1% bovine serum albumin (BSA) on the section for45min at room temperature. Sections were incubated overnightwith leucocyte and dystrophin antibodies diluted in 1% BSAat 4◦C. After overnight incubation, the slides were washedthree times for 10min in PBS-Tween in a staining jar. Slideswere moved back to the humidified chamber and sectionswere incubated for 45min with secondary antibodies diluted1:200 in 1% BSA at room temperature. Alexa Fluor R© 594F(ab′)2 fragment of goat anti-rabbit and Alexa Fluor R©488 anti-mouse IgG (Invitrogen, Eugene, Oregon, USA) were used as asecondary antibodies. The fluorochrome-stained sections werewashed three times for 10min in PBS-Tween. After the lastwashing in PBS-Tween, the sections weremounted on ProLong R©

Gold Antifade reagent with DAPI (Invitrogen, Eugene, Orgeon,USA).

Muscle sections were visualized using a high-resolutioncamera (DP72, Olympus, Japan) mounted on a microscope(BX61, Olympus, Japan) with a fluorescence light source (X-Cite 120PCQ, EXFO, Canada). The number of MPO, CD66b,and CD68 positive cells (as well as the total number of musclefibers from the area included) was counted. Data was calculatedas percentage of MPO, CD66b, or CD68 positively stained cellsper 100 skeletal muscle fibers. Areas of sections that containedfreeze damage or were folded due to the cutting procedure werenot included in the analysis.

Biochemical AssaysSerum creatine kinase activity in pre- and post-exercise bloodsamples was analyzed using an enzymatic assay (CK-NACkit, CDT14010, Thermo-Fisher Scientific Clinical Diagnostics,Sydney, Australia) and an automated clinical analyser (CobasMira, Roche Diagnostics, Germany). Serum myoglobinconcentration was also measured in these samples using animmunoassay (Roche Diagnostics, Germany) and an automatedclinical analyzer (Cobas E411, Roche Diagnostics, Germany).The intra-assay coefficient of variation was 10.4% for creatinekinase and 1.7% for myoglobin.

Muscle Soreness AssessmentUpon arrival at the laboratory and prior to the first musclebiopsy procedure, as well as 24 h following exercise prior tothe fourth and final muscle biopsy, subjects were asked to ratetheir subjective muscle soreness on a 0–10 visual analog scale.The subjects were instructed to contract, stretch, and palpatethe quadriceps muscle to assess general muscle soreness. Inboth instances 0 was considered to represent no pain andcorrespondingly a rating of 10 was considered to representintense pain.

StatisticsData are expressed as means ± SEM. Prior to analysis thedata displaying a lack of normality were log transformed tostabilize variance. Data were analyzed using a two-way ANOVAwith repeated measures for time. The sphericity adjustmentwas checked, and if required, a Greenhouse-Geisser epsiloncorrection was applied. Where no significant effect for treatmentwas observed, we explored pair-wise comparisons betweenindividual time-points using the Least Significant Difference(LSD) of means. Bivariate relationships were examined witha Spearman rank correlation test. These statistical analyseswere performed using GenStat for Windows 16th Edition (VSNInternational, Hemel Hempstead, UK). Confidence intervalswere calculated at 90% using a bootstrapping technique for non-normally distributed data using SPSS Statistics Version 22 (IBM,Portsmouth Hampshire, UK). Statistical significance was set atP < 0.05.

RESULTS

Muscle SorenessMuscle soreness showed a main effect for time (p < 0.01),suggesting that the exercise protocol was sufficient to induce amuscle stress response. No effect of ibuprofen treatment wasobserved (Figure 1).

ImmunohistochemistryThe number of MPO+ cells per 100 myofibers analyzed showed amain effect for time (P < 0.01). No main effect for treatment(P = 0.250) or time × treatment (P = 0.709) was observed(Figure 2A). LSD pairwise comparisons indicated a significantincrease in MPO+ cells at all sampling time points post-exercise.The greatest increase in MPO+ cells was observed at 3 h post-exercise (Figure 2B). Similarly, the number of CD66b+ cells per

Frontiers in Physiology | www.frontiersin.org 4 March 2016 | Volume 7 | Article 86

Vella et al. Ibuprofen and Post-exercise Inflammation

FIGURE 1 | Subjective rating for delayed onset muscle soreness

(DOMS) Values depicted are mean values ± SEM. *denotes statistical

significance from pre-exercise values (p < 0.05). White bars, PLA group; black

bars, IBU group. This figure has been adapted from Vella et al. (2014).

100 myofibers showed a main effect for time (P < 0.05), withno main effect for treatment (P = 0.149) or time × treatmentinteraction (P = 0.907; Figure 2C). LSD pairwise comparisonsindicated a significant increase in the number of CD66b+ cellsat 3 h post exercise (Figure 2D). The number of CD68+ cellsper 100 myofibers analyzed showed no effect for time (P =

0.061), treatment (P = 0.530) or time × treatment interaction(P = 0.688; Figure 2E). MPO+, CD66b+, and CD68+ cells wereobserved within the endomysium and the perimysium, with nocellular infiltration occurring at any time point. Representativeimages for each cell surface marker are presented in Figure 3.

Serum ProteinsSerum CK activity demonstrated a main effect for time (P <

0.01) with no effect for treatment (P = 0.843) or time ×

treatment interaction (P = 0.494; Figure 4A). LSD comparisonsrevealed that serum CK increased from pre-exercise values at2 h post-exercise and peaked at 24 h post-exercise (Figure 4B).Similarly, serum myoglobin concentration showed a main effectfor time (P < 0.01) with no effect for treatment (P =

0.710) or time × treatment interaction (P = 0.666; Figure 4C).LSD comparisons demonstrated that the biggest increase inserum myoglobin concentrations occurred at 1 h post-exerciseand remained significantly elevated up to 3 h post exercise(Figure 4D).

Correlation AnalysisIn general, large inter-individual differences were seen in theappearance of all of the leucocyte cell types. Results from theSpearman rank correlation test showed no correlation betweenthe histochemical appearance of inflammatory leucocytes,biological markers of muscle damage, and subjective markersof muscle soreness (Table 2). Anecdotally, three subjects wereidentified as the highest responders in the appearance of cellsurface markers applicable for localization of neutrophils (onesubject within the placebo group, and two subjects within theibuprofen group). However, these subjects showed no significantincrease in CK, myoglobin or subjective markers of muscledamage when compared with the other subjects. Interestingly,

subject 5 recorded the highest number of CD66b+ cells andMPO+ cells, whereas he presented with the lowest subjectiverating for DOMS.

DISCUSSION

Themain finding of this study was that ibuprofen, a non-selectiveCOX inhibitor, had no effect on the histological detection ofleukocytes following an acute bout of traditional resistanceexercise. Serum CK and myoglobin, and muscle sorenessalso did not change in response to ibuprofen. Furthermore,no correlation was observed between the accumulation ofinflammatory leucocytes, increases in CK or myoglobin, andsensations of DOMS. These observations suggest that theintramuscular infiltration of inflammatory white blood cells, oran increase in serum levels of intramuscular proteins, are notpredictive of post-exercise muscle soreness.

The current study focused on the acute local inflammatoryresponse to exercise-induced muscle damage. Our protocol waseffective in inducing muscle soreness as shown in Figure 1. It isimportant to note that we have previously reported this exercise-induced stress response (Vella et al., 2014). However, we felt itwas necessary to re-emphasize this response here. In the presentstudy we identified an increase in the histological appearanceof CD66b+ and MPO+ cells that peaked at 3 h post exerciseand found no effect of NSAID treatment. CD66b is a highlyspecific cell surface marker for detecting human neutrophils, andthis is the first paper to show a small but significant increasein the intramuscular number of CD66b+ cells following acuteresistance exercise. Paulsen et al. (2013) were unable to identifyany change in the number of CD66b+ cells in the elbow flexorsat 1, 2, 4, or 7 days following 70 maximal eccentric contractions(Paulsen et al., 2013). Differences in the timing of muscle biopsysamples may explain the discrepancy between the two trials, andsuggest that neutrophils may be involved in the acute phaseinflammatory response to resistance exercise. We also identifieda small but significant increase in the number of MPO+ cellsat all sampled time points, with a peak in expression at 3 hpost exercise. MPO has typically been used to detect changesin neutrophils, and an increased number of MPO+ cells havebeen reported following high-force eccentric muscle contractions(Mahoney et al., 2008; MacNeil et al., 2011). However, MPOis also expressed on the lysosomes of monocytes and has beendetected on basophils and eosinophils, perhaps providing areason why there were a higher number of MPO+ cells whencompared to CD66b+ cells detected in the present study (Paulsenet al., 2013).

No change was identified in the number of CD68+ cells. CD68cell counts are widely used as a marker of monocyte/macrophageinfiltration. However, CD68 may also be expressed on other celltypes including satellite cells and fibroblasts (Paulsen et al., 2013).Other researchers have been able to identify a change in CD68+

cells following exercise-induced muscle damage (Peterson et al.,2003; Mahoney et al., 2008; MacNeil et al., 2011). Each ofthese studies have used a high-force maximal eccentric musclecontraction protocol, which may suggest that the detection of

Frontiers in Physiology | www.frontiersin.org 5 March 2016 | Volume 7 | Article 86

Vella et al. Ibuprofen and Post-exercise Inflammation

FIGURE 2 | Immunohistochemistry data for muscle cells staining positive for myeloperoxidase (MPO), showing group specific data (A) and collapsed

data (B), CD66b showing group specific data (C), and collapsed data (D), and CD68 showing group data only (E). Data represent the mean number of

positively stained cells per 100 fibers analyzed ± SEM. **denotes statistical significance from pre-exercise values (p < 0.01). *denotes statistical significance from

pre-exercise values (p < 0.05). ∧denotes statistical significance from 0h post-exercise (p < 0.05). White bars, PLA group; black bars, IBU group.

CD68+ cells is dependent on the extent of skeletal muscledamage.

Previous research exploring the effect on NSAIDs onpost-exercise inflammation has produced equivocal findings.Earlier research from in-vivo rodent trials demonstrated ablunted inflammatory response to eccentric muscle contractionsfollowing the administration of both selective and non-selectiveNSAIDs (Lapointe et al., 2002a,b; Bondesen et al., 2004, 2006).However, these findings are yet to be supported in humantrials (Bourgeois et al., 1999; Peterson et al., 2003; Tokmakidiset al., 2003). Peterson et al. (2003) found no effect of oralibuprofen administration on the appearance of CD68+ cells 24 h

following 100 eccentric muscle contractions (Peterson et al.,2003). Similarly, Tokmakidis showed no effect of ibuprofenon circulating white blood cells 24 h following an eccentricexercise protocol (Tokmakidis et al., 2003). Paulsen et al. (2010)proposed the concept of an inflammatory threshold, suggestingthat NSAIDs may only influence post-exercise inflammation inresponse to a sufficiently strong inflammatory stimulus (Paulsenet al., 2010). It was suggested that the absence of an intramuscularprostaglandin response to exercise, specifically PGE2, couldexplain the lack of an NSAID effect on leucocyte recruitment(Trappe et al., 2001; Paulsen et al., 2010). Interestingly, ourgroup recently reported an increase in serum PGE2 in samples

Frontiers in Physiology | www.frontiersin.org 6 March 2016 | Volume 7 | Article 86

Vella et al. Ibuprofen and Post-exercise Inflammation

that were obtained during the same exercise trials as thosecurrently reported (Markworth et al., 2013). This change inserum PGE2 was blunted by the administration of ibuprofen

FIGURE 3 | Immunohistochemical analysis of skeletal muscle samples

following about of resistance exercise. Sections were probed with

antibodies raised against leucocyte cell surface markers (green; A—MPO,

B—CD66b, C—CD68) and dystrophin (red), while DAPI was used to stain

nuclei (blue). Scale bars = 50µm.

and suggests that in the presence of a blunted prostaglandinresponse to exercise, NSAID treatment had no effect on leucocyterecruitment within skeletal muscle. Although this finding doesnot refute the possibility of an inflammatory threshold, basedon our previous findings (Markworth et al., 2013), it isreasonable to suggest that the presence of an exercise-inducedserum prostaglandin response is not the required underlyingmechanism for neutrophil infiltration of muscle during the earlyhours of post-exercise recovery.

In accordance with previous studies, resistance exerciseresulted in an increase in serum CK and myoglobin, andsubjective muscle soreness (Nosaka et al., 2006). Although largeinter-individual variations in the human response to exercisecan make it difficult to establish causal relationships, therewas no relationship between the appearance of leucocytesand blood-derived markers of muscle damage or sensationsof muscle soreness. Interestingly, the subject with the highestnumber of CD66b+ and MPO+ cells presented with the lowestsubjective rating of DOMS. These findings provide support forthe assumption that leucocyte recruitment and inflammation donot consistently cause DOMS.

From animal studies it appears that cellular events in post-exercise inflammation, occur concomitantly with the onset ofDOMS (Armstrong et al., 1991; Faulkner et al., 1993). Themechanistic link has been proposed to be a leucocyte-mediated

FIGURE 4 | Blood derived proteins creatine kinase (A,B) and myoglobin (C,D). Values depicted are mean values ± SEM. (A,C) represent data divided into two

treatment groups; (B,D) represent collapsed data. *denotes statistical significance from pre-exercise values (p < 0.05). **denotes statistical significance from

pre-exercise values (p < 0.01). ∧denotes statistical significance from 1, 2, and 3 h post exercise (p < 0.05). White bars, PLA; black bars, IBU.

Frontiers in Physiology | www.frontiersin.org 7 March 2016 | Volume 7 | Article 86

Vella et al. Ibuprofen and Post-exercise Inflammation

TABLE 2 | Spearman rank correlation analysis.

CK MYO CD68+ CD66b+ MPO+ DOMS

CK – 0.59 (−0.01 − 0.91) −0.46 (−0.88 − 0.8) 0.15 (−0.42 − 0.61) 0.36 (−0.14 − 0.73) 0.01 (−0.60 − 0.62)

MYO – – 0.01 (−0.55 − 0.52) 0.14 (−0.42 − 0.69) 0.35 (−0.21 − 0.67) −0.33 (−0.74 − 0.24)

CD68+ – – – 0.05 (−0.46 − 0.56) −0.27 (−0.75 − 0.33) −0.37 (−0.82 − 0.25)

CD66b+ – – – – 0.41 (−0.13 − 0.84) −0.37 (−0.78 − 0.16)

MPO+ – – – – – 0.08 (−0.49 − 0.56)

DOMS – – – – – –

Values depicted are the correlation coefficient with 90% confidence intervals in parenthesis. Statistical significance was set at p < 0.05. No statistically significant correlations were

observed. CK, creatine kinase; MYO, myoglobin; MPO, Myeloperoxidase; DOMS, delayed-onset muscle soreness.

release of cytotoxic substances and reactive oxygen speciesas a bi-product of phagocytosis (Connolly et al., 2003).Furthermore, the prostaglandin response to exercise has beenshown to occur concurrently to the onset of DOMS, andis known to influence both the recruitment of inflammatoryleucocytes and neural afferents to pain (Markworth et al., 2013).However, in line with the current findings, the accumulationof inflammatory leucocytes in human skeletal muscle tissueduring exercise recovery appears to be both spatially andtemporally out of phase with the development of DOMS(Paulsen et al., 2010). Malm et al. (2004) explored theconcept that the activation of local leucocytes present in theepimysium may be involved in the regulation of DOMS.This hypothesis has since received support from both rodent(Gibson et al., 2009) and human trials (Crameri et al., 2007;Raastad et al., 2010) and presents a promising area for futureresearch.

CONCLUSION

This is the first study to demonstrate that ibuprofenadministration has no effect on the histological appearanceof inflammatory white blood cells following an acute boutof traditional resistance exercise. We also found no effectof ibuprofen administration on blood markers of muscledamage or subjective muscle soreness, and no significantcorrelations between leucocyte numbers and post-exercisemuscle damage or soreness. Future research needs to explorethe possibility of an inflammatory threshold above whichNSAID administration influences post-exercise inflammation.The underlying cause of post-exercise muscle soreness also

remains largely unknown. Future investigations are needed todetermine the role that inflammation plays in exercise-inducedDOMS.

AUTHOR CONTRIBUTIONS

LV—Research design, sample acquisition and analysis, dataanalysis, drafting and formatting manuscript. JM—Researchdesign, data analysis, revising manuscript, final approval ofversion to be published. GP—Data collection and analysis,revising manuscript, final approval of version to be published.TR—Data collection and analysis, interpretation of data, revising

manuscript, final approval of version to be published. JP—Research design, data analysis, interpretation of data, revisingmanuscript, final approval of version to be published. RS—Research design, interpretation of data, revising manuscript,final approval of version to be published. DC—Research design,interpretation of data, revising manuscript, final approval ofversion to be published. AR—Data analysis, interpretation ofdata, revising manuscript, final approval of version to bepublished.

ACKNOWLEDGMENTS

The authors would like to acknowledge to assistance of AssociateProfessor John Reynolds of Deakin University, and Dr. SeanWilliams of University of Bath, for their contributions to thestatistical analysis of the dataset. We further thank Dr. AndrewGarnham of Deakin University for all muscle biopsy procedures,and the participants for volunteering their time to be involved inthe trial.

REFERENCES

Armstrong, R. B.,Warren, G. L., andWarren, J. A. (1991). Mechanisms of exercise-

induced muscle fibre injury. Sports Med. 12, 184–207. doi: 10.2165/00007256-

199112030-00004

Baldwin, A. C., Stevenson, S. W., and Dudley, G. A. (2001). Nonsteroidal

anti-inflammatory therapy after eccentric exercise in healthy older

individuals. J. Gerontol. A Biol. Sci. Med. Sci. 56, M510–M513. doi:

10.1093/gerona/56.8.M510

Bondesen, B. A., Mills, S. T., Kegley, K. M., and Pavlath, G. K. (2004). The COX-2

pathway is essential during early stages of skeletal muscle regeneration. Am. J.

Physiol. Cell Physiol. 56, C475–C483. doi: 10.1152/ajpcell.00088.2004

Bondesen, B. A., Mills, S. T., and Paviath, G. K. (2006). The COX-2 pathway

regulates growth of atrophied muscle via multiple mechanisms. Am. J. Physiol.

Cell Physiol. 59, C1651–C1659. doi: 10.1152/ajpcell.00518.2005

Bourgeois, J., MacDougall, D., MacDonald, J., and Tarnopolsky, M. (1999).

Naproxen does not alter indices of muscle damage in resistance-exercise

trainedmen.Med. Sci. Sports Exerc. 31, 4–9. doi: 10.1097/00005768-199901000-

00002

Buford, T. W., Cooke, M. B., and Willoughby, D. S. (2009). Resistance exercise-

induced changes of inflammatory gene expression within human skeletal

muscle. Eur. J. Appl. Physiol. 107, 463–471. doi: 10.1007/s00421-009-1145-z

Burd, N. A., Dickinson, J. M., LeMoine, J. K., Carroll, C. C., Sullivan, B. E., Haus, J.

M., et al. (2010). Effect of a cyclooxygenase-2 inhibitor on postexercise muscle

Frontiers in Physiology | www.frontiersin.org 8 March 2016 | Volume 7 | Article 86

Vella et al. Ibuprofen and Post-exercise Inflammation

protein synthesis in humans. Am. J. Physiol. Endocrinol. Metab. 61, E354–E361.

doi: 10.1152/ajpendo.00423.2009

Connolly, D. A. J., Sayers, S. P., and McHugh, M. P. (2003). Treatment and

prevention of delayed onset muscle soreness. J. Strength Cond. Res. 17, 197–208.

doi: 10.1519/00124278-200302000-00030

Crameri, R. M., Aagaard, P., Qvortrup, K., Langberg, H., Olesen, J., and

Kjaer, M. (2007). Myofibre damage in human skeletal muscle: effects of

electrical stimulation versus voluntary contraction. J. Physiol. 583, 365–380. doi:

10.1113/jphysiol.2007.128827

Faulkner, J. A., Brooks, S. V., and Opiteck, J. A. (1993). Injury to skeletal muscle

fibers during contractions; conditions of occurrence and prevention. Phys.

Ther. 73, 911–921.

Fridén, J., Sjöström, M., and Ekblom, B. (1981). A morphological study of delayed

muscle soreness. Experientia 37, 506–507. doi: 10.1007/BF01986165

Gibson,W., Arendt-Nielsen, L., Taguchi, T., Mizumura, K., andGraven-Nielsen, T.

(2009). Increased pain from muscle fascia following eccentric exercise: animal

and human findings. Exp. Brain Res. 194, 299–308. doi: 10.1007/s00221-008-

1699-8

Hersh, E. V., Moore, P. A., and Ross, G. L. (2000). Over-the-counter analgesics and

antipyretics: a critical assessment. Clin. Ther. 22, 500–548. doi: 10.1016/S0149-

2918(00)80043-0

Hyldahl, R. D., Keadle, J., Rourier, P. A., Pearl, D., and Clarkson, P. M. (2010).

Effects of ibuprofen topical gel on muscle soreness. Med. Sci. Sports Exerc. 42,

614–621. doi: 10.1249/MSS.0b013e3181b95db2

Krentz, J. R., Quest, B., Farthing, J. P., Quest, D. W., and Chilibeck, P. D. (2008).

The effects of ibuprofen on muscle hypertrophy, strength, and soreness during

resistance training. Appl. Physiol. Nutr. Metab. 33, 470–475. doi: 10.1139/H08-

019

Lapointe, B. M., Frémont, P., and Côté, C. H. (2003). Influence of nonsteroidal

anti-inflammatory drug treatment duration and time of onset on recovery from

exercise-induced muscle damage in rats. Arch. Phys. Med. Rehabil. 84, 651–655.

doi: 10.1016/S0003-9993(02)04899-2

Lapointe, B. M., Frémont, P., and Côté, C. H. (2002a). Adaptation to lengthening

contractions is independent of voluntary muscle recruitment but relies on

inflammation. Am. J. Physiol. Regul. Integr. Comp. Physiol. 282, R323–R329.

doi: 10.1152/ajpregu.00339.2001

Lapointe, B. M., Frenette, J., and Côté, C. H. (2002b). Lengthening contraction-

induced inflammation is linked to secondary damage but devoid of neutrophil

invasion. J. Appl. Physiol. 92, 1995–2004. doi: 10.1152/japplphysiol.00803.2001

MacNeil, L. G., Baker, S. K., Stevic, I., and Tarnopolsky, M. A. (2011).

17β-estradiol attenuates exercise-induced neutrophil infiltration in men.

Am. J. Physiol. Regul. Integr. Comp. Physiol. 300, R1443–R1451. doi:

10.1152/ajpregu.00689.2009

Mahoney, D. J., Safdar, A., Parise, G., Melov, S., Fu, M., MacNeil, L., et al.

(2008). Gene expression profiling in human skeletal muscle during recovery

from eccentric exercise. Am. J. Physiol. Regul. Integr. Comp. Physiol. 294,

R1901–R1910. doi: 10.1152/ajpregu.00847.2007

Malm, C., Sjödin, T. L. B., Sjöberg, B., Lenkei, R., Renström, P., Lundberg, I. E., et al.

(2004). Leukocytes, cytokines, growth factors and hormones in human skeletal

muscle and blood after uphill or downhill running. J. Physiol. 556, 983–1000.

doi: 10.1113/jphysiol.2003.056598

Markworth, J. F., Vella, L. D., Figueiredo, V. C., and Cameron-Smith, D. (2014).

Ibuprofen treatment blunts early translational signaling responses in human

skeletal muscle following resistance exercise. J. Appl. Physiol. 117, 20–28. doi:

10.1152/japplphysiol.01299.2013

Markworth, J. F., Vella, L., Lingard, B. S., Tull, D. L., Rupasinghe, T.W., Sinclair, A.

J., et al. (2013). Human inflammatory and resolving lipid mediator responses to

resistance exercise and ibuprofen treatment.Am. J. Physiol. Regul. Integr. Comp.

Physiol. 305, R1281–R1296. doi: 10.1152/ajpregu.00128.2013

Mikkelsen, U. R., Langberg, H., Helmark, I. C., Skovgaard, D., Andersen, L. L.,

Kjaer, M., et al. (2009). Local NSAID infusion inhibits satellite cell proliferation

in human skeletal muscle after eccentric exercise. J. Appl. Physiol. 107,

1600–1611. doi: 10.1152/japplphysiol.00707.2009

Mikkelsen, U. R., Schjerling, P., Helmark, I. C., Reitelseder, S., Holm, L.,

Skovgaard, D., et al. (2011). Local NSAID infusion does not affect protein

synthesis and gene expression in human muscle after eccentric exercise.

Scand. J. Med. Sci. Sports 21, 630–644. doi: 10.1111/j.1600-0838.2010.

01170.x

Mishra, D. K., Fridén, J., Schmitz, M. C., and Lieber, R. L. (1995). Anti-

inflammatory medication after muscle injury. A treatment resulting in short-

term improvement but subsequent loss of muscle function. J. Bone Joint Surg.

Am. 77, 1510–1519.

Nosaka, K., Sacco, P., and Mawatari, K. (2006). Effects of amino acid

supplementation on muscle soreness and damage. Int. J. Sport Nutr. Exerc.

Metab. 16, 620–635.

Paulsen, G., Egner, I. M., Drange, M., Langberg, H., Benestad, H. B., Fjeld, J. G.,

et al. (2010). A COX-2 inhibitor reducesmuscle soreness, but does not influence

recovery and adaptation after eccentric exercise. Scand. J. Med. Sci. Sports 20,

1–13. doi: 10.1111/j.1600-0838.2009.00947.x

Paulsen, G., Egner, I., Raastad, T., Reinholt, F., Owe, S., Lauritzen, F., et al. (2013).

Inflammatory markers CD11b, CD16, CD66b, CD68, myeloperoxidase and

neutrophil elastase in eccentric exercised human skeletal muscles. Histochem.

Cell Biol. 139, 691–715. doi: 10.1007/s00418-012-1061-x

Peterson, J. M., Trappe, T. A., Mylona, E., White, F., Lambert, C. P., Evans,

W. J., et al. (2003). Ibuprofen and acetaminophen: effect on muscle

inflammation after eccentric exercise. Med. Sci. Sports Exerc. 35, 892–896. doi:

10.1249/01.MSS.0000069917.51742.98

Raastad, T., Owe, S. G., Paulsen, G., Enns, D., Overgaard, K., Crameri,

R., et al. (2010). Changes in calpain activity, muscle structure, and

function after eccentric exercise. Med. Sci. Sports Exerc. 42, 86–95. doi:

10.1249/MSS.0b013e3181ac7afa

Smith, L. L. (1991). Acute inflammation: the underlying mechanism in

delayed onset muscle soreness. Med. Sci. Sports Exerc. 23, 542–551. doi:

10.1249/00005768-199105000-00006

Tidball, J. G. (1995). Inflammatory cell response to acute muscle injury.

Med. Sci. Sports Exerc. 27, 1022–1032. doi: 10.1249/00005768-199507000-

00011

Tokmakidis, S. P., Kokkinidis, E. A., Smilios, I., and Douda, H. (2003). The effects

of ibuprofen on delayed muscle soreness and muscular performance after

eccentric exercise. J. Strength Condition. Res. 17, 53–59. doi: 10.1519/00124278-

200302000-00009

Trappe, T. A., Fluckey, J. D., White, F., Lambert, C. P., and Evans, W. J. (2001).

Skeletal muscle PGF(2)(alpha) and PGE(2) in response to eccentric resistance

exercise: influence of ibuprofen acetaminophen. J. Clin. Endocrinol. Metab. 86,

5067–5070. doi: 10.1210/jcem.86.10.7928

Trappe, T. A., White, F., Lambert, C. P., Cesar, D., Hellerstein, M., and Evans,

W. J. (2002). Effect of ibuprofen and acetaminophen on postexercise muscle

protein synthesis. Am. J. Physiol. Endocrinol. Metab. 282, E551–E556. doi:

10.1152/ajpendo.00352.2001

Urso, M. L. (2013). Anti-inflammatory interventions and skeletal muscle

injury: benefit or detriment? J. Appl. Physiol. 115, 920–928. doi:

10.1152/japplphysiol.00036.2013

Vella, L., Caldow, M. K., Larsen, A. E., Tassoni, D., Della Gatta, P. A., Gran, P.,

et al. (2012). Resistance exercise increases NF-κB activity in human skeletal

muscle. Am. J. Physiol. Regul. Integr. Comp. Physiol. 302, R667–R673. doi:

10.1152/ajpregu.00336.2011

Vella, L., Markworth, J. F., Peake, J. M., Snow, R. J., Cameron-Smith, D., and

Russell, A. P. (2014). Ibuprofen supplementation and its effects on NF-

κB activation in skeletal muscle following resistance exercise. Physiol. Rep.

2:e12172. doi: 10.14814/phy2.12172

Whisenant, M. J., Panton, L. B., East, W. B., and Broeder, C. E. (2003). Validation

of submaximal prediction equations for the 1 repetition maximum bench press

test on a group of collegiate football players. J. Strength Condition. Res. 17,

221–227. doi: 10.1519/00124278-200305000-00002

Conflict of Interest Statement: The authors declare that the research was

conducted in the absence of any commercial or financial relationships that could

be construed as a potential conflict of interest.

Copyright © 2016 Vella, Markworth, Paulsen, Raastad, Peake, Snow, Cameron-

Smith and Russell. This is an open-access article distributed under the terms

of the Creative Commons Attribution License (CC BY). The use, distribution or

reproduction in other forums is permitted, provided the original author(s) or licensor

are credited and that the original publication in this journal is cited, in accordance

with accepted academic practice. No use, distribution or reproduction is permitted

which does not comply with these terms.

Frontiers in Physiology | www.frontiersin.org 9 March 2016 | Volume 7 | Article 86

131

Appendix B:

ORIGINAL RESEARCH

Ibuprofen supplementation and its effects on NF-jBactivation in skeletal muscle following resistance exerciseLuke Vella1, James F. Markworth2, Jonathan M. Peake3, Rod J. Snow1, David Cameron-Smith2 &Aaron P. Russell1

1 Centre for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Burwood, Vic., Australia

2 Liggins Institute, University of Auckland, Auckland, New Zealand

3 School of Biomedical Sciences and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Qld, Australia

Keywords

Exercise recovery, inflammation, NF-jB,

NSAID treatment, resistance exercise.

Correspondence

Luke Vella, Centre for Physical Activity and

Nutrition, School of Exercise and Nutrition

Science, Deakin University, 221 Burwood

Highway, Burwood, Victoria 3125, Australia.

Tel: (03) 9244 6100

Fax: (03) 9786 0141

E-mail: [email protected]

Funding Information

No funding information provided.

Received: 10 September 2014; Accepted: 10

September 2014

doi: 10.14814/phy2.12172

Physiol Rep, 2 (10), 2014, e12172,

doi: 10.14814/phy2.12172

Abstract

Resistance exercise triggers a subclinical inflammatory response that plays a

pivotal role in skeletal muscle regeneration. Nuclear factor-jB (NF-jB) is a

stress signalling transcription factor that regulates acute and chronic states of

inflammation. The classical NF-jB pathway regulates the early activation of

post-exercise inflammation; however there remains scope for this complex

transcription factor to play a more detailed role in post-exercise muscle recov-

ery. Sixteen volunteers completed a bout of lower body resistance exercise

with the ingestion of three 400 mg doses of ibuprofen or a placebo control.

Muscle biopsy samples were obtained prior to exercise and at 0, 3 and 24 h

post-exercise and analysed for key markers of NF-jB activity. Phosphorylated

p65 protein expression and p65 inflammatory target genes were elevated

immediately post-exercise independent of the two treatments. These changes

did not translate to an increase in p65 DNA binding activity. NF-jB p50 pro-

tein expression and NF-jB p50 binding activity were lower than pre-exercise

at 0 and 3 h post-exercise, but were elevated at 24 h post-exercise. These find-

ings provide novel evidence that two distinct NF-jB pathways are active in

skeletal muscle after resistance exercise. The initial wave of activity involving

p65 resembles the classical pathway and is associated with the onset of an

acute inflammatory response. The second wave of NF-jB activity comprises

the p50 subunit, which has been previously shown to resolve an acute inflam-

matory program. The current study showed no effect of the ibuprofen treat-

ment on markers of the NF-jB pathway, however examination of the within

group effects of the exercise protocol suggests that this pathway warrants

further research.

Introduction

Unaccustomed resistance exercise causes skeletal muscle

damage that impairs muscle function and promotes sen-

sations of pain. The precise signalling mechanisms that

initiate muscle repair following exercise-induced damage

remain a topic of ongoing debate [previously reviewed by

Paulsen et al. (2012)]. The onset of muscle damage trig-

gers a complex interplay of intracellular events that

involve myofibre injury, acute inflammation and cellular

repair. At a symptomatic level, inflammation in skeletal

muscle is characterised by swelling and soreness (Paulsen

et al. 2012). Consequently post-exercise inflammation is

often ascribed as a cause of delayed-onset muscle soreness

(DOMS). DOMS typically occurs 24–48 h post-exercise,

and is accompanied by a secondary reduction in muscle

force-generating capacity. Treatment of DOMS has

focused on reducing inflammation; however this may be

detrimental to processes of cellular repair (Urso 2013).

Attenuating exercise-induced inflammation using non-ste-

roidal anti-inflammatory drugs reduces skeletal muscle

protein synthesis (Trappe et al. 2002) and impairs satellite

cell proliferation (Mikkelsen et al. 2009). To develop

more efficacious treatments for DOMS, a better

ª 2014 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of

the American Physiological Society and The Physiological Society.

This is an open access article under the terms of the Creative Commons Attribution License,

which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

2014 | Vol. 2 | Iss. 10 | e12172Page 1

Physiological Reports ISSN 2051-817X

understanding of the mechanisms that govern inflamma-

tion and tissue repair in skeletal muscle after exercise is

required.

The nuclear factor-kappa B (NF-jB) transcription

factor acts as a central regulator of inflammatory signal-

ling pathways. The NF-jB family consists of five subunits,

including NF-jB1 (p105/p50), NF-jB2 (p100/p52), RelA

(p65), RelB and cREL that share an amino terminus Rel

homology domain. The Rel domain permits DNA bind-

ing, nuclear localization, dimerization and interaction

with its own inhibitory protein IjB (Delhalle et al. 2004;

Mourkioti and Rosenthal 2008; Bakkar and Guttridge

2010). Under basal conditions RelA (p65), cREL and RelB

subunits remain sequestered within the cytoplasm bound

to an inhibitory IjB protein. The IjB family that regu-

lates NF-jB includes the subunits IjBb, IjBa, IjBc IjBe,and Bcl-3. Unlike the Rel proteins, subunits p50 and p52

are synthesized as large precursor proteins (p105 and 100,

respectively) that require proteolytic processing to permit

nuclear localization. These subunits lack a REL domain,

to initiate gene transcription, and hence are primarily

considered as repressors of gene transcription (Bonizzi

and Karin 2004; Bakkar and Guttridge 2010).

Upon stimulation, the IjB kinase complex (IKK) con-

trols the degradation of IjB and its precursor proteins,

thereby enabling the NF-jB REL subunits to control gene

transcription. The IKK complex consists of two catalytic

kinases IKKa and IKKb, and a regulatory IKKc subunit.

IKKb activates the classical NF-jB signalling pathway

through the phosphorylation and subsequent degradation

of the inhibitor IjBa (Zandi et al. 1997; Pahl 1999). The

classical pathway typically comprises p65:p50 heterodi-

mers, and is essential for the activation of acute inflam-

mation by controlling the transcription on inflammatory

cytokines and acute phase proteins (Zandi et al. 1997;

Pahl 1999). More recently an alternative NF-jB pathway

has been described, which is dependent on IKKa (Senftle-

ben et al. 2001). Alternative signalling involves the activa-

tion of a secondary NF-jB inducing kinase (NIK), and

has been linked to the activation of both p52 and p50

subunits. The functional significance of IKKa-dependentgene expression in acute inflammation is not yet well

established. However, preliminary research in rat muscle

tissue suggests p50 homodimers may play a crucial role

in the resolution of acute inflammation (Bohuslav et al.

1998; Lawrence et al. 2001).

Despite the importance of inflammation in tissue

regeneration post-exercise, very little is known about how

NF-jB activity is regulated in skeletal muscle after acute

exercise. A transient increase in various components of

the classical NF-jB signalling pathway is observed in rat

muscle post-exercise (Hollander et al. 2001; Ji et al. 2004;

Ho et al. 2005; Spangenburg et al. 2006; Kramer and

Goodyear 2007). In contrast, a decrease in NF-jB DNA

binding at 0 h post-exercise with a return to near baseline

1 h post-exercise was observed in human muscle follow-

ing a traditional resistance exercise model (Durham et al.

2004). Recent work from our laboratory, using a similar

resistance exercise model, demonstrated an increase in

NF-jB binding to the promoter region of key inflamma-

tory cytokines at 2 h post-exercise, with a return to base-

line levels at 4 h post-exercise (Vella et al. 2012). These

findings suggest that a transient NF-jB response contrib-

utes to acute post-exercise inflammation.

To enhance our understanding of the cellular mecha-

nisms that regulate both the onset and resolution of post-

exercise inflammation, the current study aimed to investi-

gate changes in the activity of the subunits that comprise

the classical and alternative NF-jB signalling pathways.

We hypothesized that the regulation of NF-jB post-exer-

cise would involve two distinct waves of activation. Spe-

cifically, we hypothesized that the classical NF-jBpathway, involving p65 and p50 dimers would be acti-

vated soon after exercise during the early phases of

inflammation, while the alternative pathway, comprising

mainly the p50 subunit would be activated at later time

points after exercise that correspond with the resolution

of acute inflammation (Bohuslav et al. 1998; Ishikawa

et al. 1998). We also hypothesized that the administration

of ibuprofen would blunt both waves of NF-jB activa-

tion, providing a potential mechanism through which

anti-inflammatory medication might attenuate exercise-

induced inflammation in skeletal muscle.

Methods

Participants

As previously described, sixteen healthy male subjects

were recruited to participate in the study (characteristics

shown in Table 1) (Markworth et al. 2013). All partici-

pants completed a medical history questionnaire that was

used to identify and exclude participants with a diagnosed

condition or illness that prevented them from completing

strenuous exercise. Exclusion criteria included participa-

tion in a lower body resistance exercise program within

the last 6 months to ensure a muscle damage response

from the exercise stimulus, and/or chronic treatment with

anti-inflammatory drugs. Current use of prescription

medication or nutritional supplements also excluded sub-

jects from participating.

Ethics approval

Prior to participation each subject received written and

oral information regarding the nature of the experiment

2014 | Vol. 2 | Iss. 10 | e12172Page 2

ª 2014 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of

the American Physiological Society and The Physiological Society.

NF-jB and Resistance Exercise L. Vella et al.

before providing written consent to participate. All proce-

dures involved in this study were approved by the Deakin

University Human Research Ethics Committee (DUHREC

2010-019). All muscle sampling procedures were per-

formed in accordance with the Helsinki declaration.

Familiarization and strength testing

Each participant completed a familiarization session at

least 7 days prior to completing the exercise trial. Partici-

pants performed repetition maximum testing to deter-

mine the experimental exercise load (80% of 1 repetition

maximum (1RM)). The maximal weight each subject

could lift was determined for the Smith machine-assisted

squat, the leg press, and the leg extension. These data

were substituted into the validated Brzycki equation to

predict 1RM for each participant (Mayhew et al. 1995;

Whisenant et al. 2003). The participants were required to

abstain from any further exercise until completion of the

trial.

Experimental Procedures

The participants reported to the laboratory in an over-

night fasted state, having abstained from caffeine, tobacco

and alcohol for the preceding 24 h. Following 30 min of

supine resting a pre-exercise muscle biopsy was taken.

Participants then completed a 10 min warm up consisting

of low intensity cycling on a bicycle ergometer, and one

low resistance set for each exercise at approximately 30–50% of the participants 1RM. Each participant then com-

pleted a single bout of intense resistance exercise. This

session consisted of three sets of 8–10 repetitions of a

bilateral Smith machine assisted squat, 45° leg press and

leg extension. These exercises were all performed at 80%

1RM. The exercises were performed sequentially as a cir-

cuit, with 1 min rest between each exercise, and 3 min

rest between sets. We have previously used this exercise

protocol and demonstrated that it activates inflammatory

signalling pathways (Vella et al. 2012). Following the

completion of the exercise bout, the participants rested in

a supine position while muscle biopsy samples were col-

lected. The participants returned to the laboratory the fol-

lowing morning in an over-night fasted state for a final

muscle biopsy sample. Standardized meals were provided

to participants the night preceding the trial (carbohydrate

57%, fat 22%, protein 21%), in the laboratory immedi-

ately following the exercise bout (carbohydrate 71%, fat

13%, protein 16%), as additional snacks throughout the

day and an evening meal (carbohydrate 64%, fat 27%,

protein 18%).

NSAID administration

Prior to the exercise bout, the participants were randomly

assigned to either the ibuprofen (NSAID) group (n = 8)

or the placebo group (PLA) (n = 8). Participants in the

NSAID group consumed the maximum recommended

over-the-counter dose of 1200 mg of ibuprofen as three

doses of 400 mg throughout the trial day. The first dose

was administered upon arriving to the laboratory on the

first morning of the trial, immediately prior to the first

muscle biopsy sample. Participants were instructed to

consume two additional doses at 2:00 pm and 8:00 pm

the same evening. This dosing structure was prescribed to

optimise levels of circulating ibuprofen to biologically

active levels throughout the course of the trial day. This

protocol has previously been validated by our research

group in this same group of study participants and the

same NSAID administration protocol (Markworth et al.

2013). Alternatively the placebo consumed a gelatin cap-

sule containing powdered sugar, identical in appearance

to the ibuprofen capsule.

Sample collection

Muscle biopsy samples were obtained under local anaes-

thesia (Xylocaine 1%) from the vastus lateralis muscle of

either leg using the percutaneous needle biopsy technique

modified to include suction (Buford et al. 2009). Samples

were obtained prior to exercise, immediately following

exercise and at 3 and 24 h post-exercise. The muscle

biopsy sample obtained immediately post-exercise was

taken within 1–2 min of the completion of the exercise

protocol, and will herein after be referred to as 0 h post-

exercise. The muscle biopsy procedure has been shown to

Table 1. Subject characteristics and strength testing data.

Characteristics Strength (1RM)

Age (y) Height (m) Body mass (kg) BMI Squat(kg) Leg press (kg) Leg extension (kg)

PLA 23.9 � 1.3 1.89 � 0.1 86.9 � 4.5 24.5 � 1.2 94.9 � 5.4 237 � 17 236 � 18

IBU 23.0 � 0.5 1.89 � 0.1 89.1 � 4.4 24.8 � 0.8 91.9 � 6.0 240 � 15 196 � 22

Values are mean values � SEM. No significant differences were observed between the two groups.

ª 2014 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf ofthe American Physiological Society and The Physiological Society.

2014 | Vol. 2 | Iss. 10 | e12172Page 3

L. Vella et al. NF-jB and Resistance Exercise

trigger a local inflammatory response (Malm et al. 2000).

To minimize interference from the biopsy procedure,

samples prior to exercise and at 24 h post-exercise were

taken from the same leg, while muscle samples obtained

at 0 and 3 h post-exercise were taken from the opposite

leg. Samples within the same leg were taken at least 5 cm

from the previous site. We have previously reported that

this technique is effective for minimizing cytokine gene

expression and NF-jB activity in response to muscle

biopsies (Vella et al. 2012). Excised tissue was immedi-

ately immersed in liquid nitrogen and stored at �80°Cuntil further analysis.

Subjective assessment of DOMS and rangeof movement

Subjective assessment of muscle soreness (DOMS) and

range of movement were recorded prior to exercise and

at 24 h post exercise. Subjects were asked to rate their

levels of muscle soreness and range of movement on a

0–10 scale. In both instances 0 was considered as the best

possible result and a rating of 10 was considered to be

the worst result.

Protein extraction and quantification

Muscle samples were homogenized in ice cold RIPA buf-

fer (50 mmol/L Tris-HCl, pH 7.4, 150 mmol/L NaCl,

0.25% deoxycholic acid, 1% NP-40, 1 mmol/L EDTA

supplemented with protease and phosphatase inhibitors

including 1 mmol/L PMSF, 1 lg/mL aprotinin, 1 lg/mL

leupeptin, 1 mmol/L Na3VO4 & 1 mmol/L NaF). The

homogenate was agitated for 1 h at 4°C and centrifuged

at 13,000 9 g at 4°C for 15 min. The resultant superna-

tant was removed and stored at �80°C until further

analysis. Total protein concentration was determined

using a BCA protein assay kit according to the manufac-

turer’s instruction (Pierce, Rockford, IL). Protein samples

(50 lg) were denatured in loading buffer and separated

by a 10% SDS-PAGE and transferred to a PVDF mem-

brane. Membranes were blocked for 90 min at room

temperature in 5% BSA/Tris buffered saline with 0.1%

Tween 20 (TBST). Primary antibodies [phosphorylated

NF-jB p65 Ser536, total NF-jB p65, NF-jB p100/p52,

NF-jB p105/p50, NF-jB cREL and b-actin (all obtained

from Cell Signaling Technologies, Arundel, QLD)] were

diluted to 1:1000 in 5% BSA/TBST, applied and incu-

bated overnight at 4°C with gentle agitation. Membranes

were washed for 30 min in TBST and probed with HRP

conjugated secondary antibodies diluted to 1:2000 in 5%

BSA/TBST, and incubated for 1 h at room temperature.

Proteins were visualised by using Western Lighting

enhanced chemiluminescence (Perkin Elmer Lifesciences,

Boston, MA). Signals were captured using a Kodak Digi-

tal Image Station 2000M (model: 440CF; Eastman Kodak,

Rochester, NY) and quantified by densitometry band

analysis using Kodak Molecular Imaging Software (Ver-

sion 4.0.5, © 1994–2005, Eastman Kodak). Phosphory-

lated NF-jB p65 protein was normalized to total p65

protein (Fig. 2A). NF-jB p50 protein expression was

normalized to its precursor protein p105 (Fig. 2B). NF-

jB p52 and cREL protein was normalized to b-actin(Fig. 2C).

RNA extraction and RT-PCR

Total cellular RNA was extracted as previously described

(Trenerry et al. 2007) using the ToTALLY RNA Kit (Am-

bion, Austin, TX). RNA quality and concentration were

determined using the Agilent 2100 Biolanalyzer (Agilent

Technologies, Palo Alto, CA). First strand cDNA was gen-

erated from 0.5 lg total RNA using the AMV RT kit

(Promega, Madison, WI). RT-PCR was performed in

duplicate using the Biorad CFX384 system (Biorad, Her-

cules, CA), containing 5XHOT FirePol� EvaGreen Mix

(Integrated Science, Sydney, NSW), forward primer,

reverse primer, sterile nuclease free water and cDNA

(0.125 ng/lL). Data were analysed using a comparative

critical threshold (Ct) method, where the amount of the

specified target gene normalised to the amount of endog-

enous control, relative to the control value is given by

2�DDCt . The endogenous control used in this experiment

was GAPDH. The efficacy of GAPDH as an endogenous

control was examined using the equation 2�DCt . Primers

were designed using Primer Express software package ver-

sion 3.0 (Applied Biosystems, Mulgrave, Vic., Australia).

Gene sequences were obtained from GenBank (Table 2).

Primer sequence specificity was confirmed using BLAST.

A melting point dissociation curve was generated by the

PCR instrument for all PCR products to confirm the

presence of a single amplified product.

Nuclear extraction and Transcription Factor(TF) Assay

Nuclear and cytoplasmic proteins were extracted from

20 mg of muscle tissue using a NE-PER Nuclear and

Cytoplasmic Extractions Reagents (Pierce, Rockford, IL),

according to the manufacturer’s instructions. Western

blot analysis probed for GAPDH, a-tubulin and Lamin A

was performed to ensure the nuclear extract was not

contaminated by cytoplasmic proteins. A Transcription

Factor Assay detecting specific transcription factor

DNA binding activity was performed according to

manufacturer’s instructions (Cayman Chemical, Ann

Arbour, MI). Briefly, 10 lg of nuclear protein were

2014 | Vol. 2 | Iss. 10 | e12172Page 4

ª 2014 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of

the American Physiological Society and The Physiological Society.

NF-jB and Resistance Exercise L. Vella et al.

loaded into a well containing an immobilized NF-jB con-

sensus sequence (50GGGACTTTCC-30) and incubated

overnight at 4°C. Primary antibodies for NF-jB subunits

p65 and p50 were loaded in to each well and incubated at

room temperature for 1 h. Each well was flushed using a

diluted wash buffer for 30 min. Following a secondary

30 min wash, samples were incubated with secondary

HRP-conjugated antibody for a further 1 h at room tem-

perature. To quantify transcription factor binding, a

developing solution containing a 3,30,5,50-Tetrame-

thhylbenzidine (TMB) solution was added to each

well and incubated for 45 min at room temperature.

DNA binding was then quantified using photospectrome-

try with absorbance measurements taken at 450 nm

using a Multiscan RC plate reader (Labsystems, Finland)

and Gen5 Data Analysis Software (BioTek, Winooski,

VT).

Statistics

Data are expressed as means � SEM. Prior to analysis the

data displaying a lack of normality were log transformed

to stabilise variance. Data were analysed using a two-way

ANOVA with repeated measures for time. The sphericity

adjustment was checked, and if required, a Greenhouse-

Geisser epsilon to the residual degrees of freedom was

applied. A data point was considered to be a statistical

outlier where a z-score exceeded a pre-determined thresh-

old of � 4.00. Where appropriate we explored within-

group pair-wise comparisons between individual time-

points using the Least Significant Differences of means at

P < 0.05 to determine statistically significant changes

(Mead 1988; Saville 1990). Statistical analyses were per-

formed using GenStat for Windows 16th Edition (VSN

International, Hemel Hempstead, UK).

Results

Subjective DOMS analysis showed a main effect for time

(time effect P < 0.01) with no effect for treatment. The

average post-exercise DOMS score was 4.81 � 0.5 and

the post-exercise ROM score was 3.3 � 0.5. This data

demonstrates that the exercise protocol induced a moder-

ate level of muscle soreness and this effect was irrespective

of ibuprofen treatment.

Expression of NF-jB phosphorylated p65 (Ser 536)

increased over time (time effect P = 0.006), but this

response was overall not different between the groups

(interaction effect P = 0.253, treatment effect P = 0.176).

Nevertheless, LSD pairwise comparisons indicated that

the main increase from baseline was in the placebo

group at 0 h post-exercise. Within the placebo group

phosphorylated p65 protein expression remained elevated

at 3 h post-exercise and returned to baseline by 24 h

(Fig. 1A). No significant changes were observed in the

ibuprofen group by LSD comparisons. There was no

effect of time or treatment for NF-jB total p65

(Fig. 2A).

There was a trend toward a significant time 9 treat-

ment interaction (P = 0.057) for the protein expression

of the p50 subunit, although a main effect for time

(P = 0.376) or treatment (P = 0.865) was not achieved.

Our analysis revealed a statistical outlier in the ibuprofen

group at the 24 h time point. When this subject was

removed from the analysis, this trend was weaker

(P = 0.115) (Fig. 1B). LSD comparisons indicated an

increase in p50 protein expression was observed at 24 h

post exercise in the placebo group only (Fig. 1B). There

were no main or interaction effects for the NF-jB p50

precursor protein p105 (Fig. 2B) or for p52 and cREL

subunits (Fig. 1C and D).

To determine subunit-specific DNA binding of NF-jBfollowing exercise, transcription factor binding assays

were performed for p50 and p65 subunits. NF-jB p50

showed a main effect for time (P = 0.035), with no

time 9 treatment interaction (P = 0.851) or main effect

for treatment (P = 0.488) (Fig. 3B). LSD comparisons

identified a significant increase in p50 binding that

occurred at 24 h post-exercise when compared to 0 and

3 h post-exercise within the placebo group. This coin-

cided with the increase in p50 protein expression

(Fig. 1B). No change in p65 DNA binding was observed,

despite an increase in phosphorylated p65 protein expres-

sion (Fig. 3B).

We sought to determine whether any increase in

NF-jB signalling coincided with an increase in down-

stream inflammatory cytokines, and if the administration

of ibuprofen influenced post-exercise cytokine expression.

The mRNA levels of IL-6 (Fig. 4A), IL-8 (Fig. 4B) and

MCP-1 (Fig. 4C) demonstrated a main effect for time

(P < 0.01), with the highest elevation in expression levels

at 3 h post-exercise after significant increases at 0 h post-

exercise. TNF-a mRNA remained unchanged after exer-

cise (Fig. 4D). COX-2 mRNA showed a main effect for

time (P < 0.01), increasing at 0, 3 and 24 h post-exercise

(Fig. 4E). There were no significant interaction effects or

main effects for treatment for the inflammatory cytokines

or COX-2 mRNA expression. Consistently, LSD pairwise

comparisons revealed similar changes over time from

baseline within both groups.

Discussion

The current study aimed to explore the regulation of

the different NF-jB subunits following a single bout of

lower body resistance exercise, and investigate the mech-

ª 2014 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf ofthe American Physiological Society and The Physiological Society.

2014 | Vol. 2 | Iss. 10 | e12172Page 5

L. Vella et al. NF-jB and Resistance Exercise

anism through which ibuprofen treatment may influence

post-exercise inflammation. The results of this study

show for the first time that an alternative NF-jB signal-

ling pathway comprising mainly p50 subunits is acti-

vated 24 h post-exercise. This pathway appears distinctly

different to the activation of NF-jB p65 subunit that

occurs during the early stages of acute post-exercise

inflammation.

Phosphorylated NF-jB p65 protein expression was sig-

nificantly elevated in the placebo group when measured

at 0 and 3 h post-exercise, and returned to baseline levels

at 24 h post-exercise. These findings support those of pre-

vious research showing a post-exercise increase in key

components of the classical NF-jB signalling pathway in

skeletal muscle, including phosphorylated NF-jB p65

protein (Ji et al. 2004; Vella et al. 2012), phosphorylated

IjBa protein (Ho et al. 2005; Vella et al. 2012) and NF-

jB p65 DNA binding activity (Tantiwong et al. 2010;

Hyldahl et al. 2011). In the present study, the increase in

phosphorylated NF-jB p65 protein expression was not

associated with an increase in p65 transcription factor

binding activity at 0, 3 and 24 h post-exercise. However,

NF-jB regulated cytokine genes including MCP-1, IL-6

and IL-8 were increased at 3 h post-exercise. Previous

work from our group showed an increase in NF-jB p65

binding to the promoter region of genes coding for

inflammatory cytokines at 2 h after traditional resistance

exercise (Vella et al. 2012). This research performed a ser-

ies of electrophoretic mobility shift assays that looked

specifically NF-jB binding to genes coding MCP-1, IL-6

and IL-8 and thus differences in the methodology may

explain conflicting results. Furthermore, the discrepancy

in the results between our two trials may be explained by

the short half-life of NF-jB in the absence of an activat-

ing stimulus. In a HL60 cell line, the half-life of NF-jBhas been reported to be less than 30 min (Hohmann

et al. 1991). Likewise, the inhibitory IjBa protein has a

half-life of 25 min in Jurkat cells (Dodd et al. 2009).

Therefore, in the present study, NF-jB activation may

have returned to resting levels by 3 h post-exercise.

Research models using an extreme eccentric resistance

exercise model were able to demonstrate an increase in

p65 DNA binding to nuclear protein at 3 h post-exercise

(Hyldahl et al. 2011; Xin et al. 2013). By contrast,

A B

C D

Figure 1. Protein expression of NF-jB subunits p65, p50, p52 and cREL. Representative Western blots for p-p65 normalized to total p65

(A), p50 normalized to p105 (B), p52 normalized to b-actin (C) and cREL normalized to b-actin (D) measured in muscle biopsy samples. Data

are mean arbitrary units � SEM. *denotes statistical significance from pre exercise values in the placebo group; ^denotes statistical significance

from 24 h post-exercise in the control group (P < 0.05). White bars = placebo group; black bars = ibuprofen group.

2014 | Vol. 2 | Iss. 10 | e12172Page 6

ª 2014 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of

the American Physiological Society and The Physiological Society.

NF-jB and Resistance Exercise L. Vella et al.

research from Durham et al. (2004) showed a decrease in

NF-jB DNA binding activity immediately post-exercise,

which returned to baseline levels at 1 h post-exercise.

Collectively, these findings suggest that the activation of

NF-jB DNA binding occurs transiently within the first

few hours of exercise. Changes in NF-jB activity in skele-

tal muscle may depend on the mode of exercise and

training status of participants.

This research also provides novel evidence for a

potential role of NF-jB p50 activation as a component

of inflammation-resolution. In our initial analysis of

p50 protein expression, there was a trend towards a

time 9 group interaction effect (P = 0.057) but this

trend was weaker (P = 0.115) after removing a statistical

outlier. Despite this weaker interaction, within-group

analysis by LSD comparisons revealed that p50 protein

A B

C

Figure 2. Protein expression of total NF-jB p65, NF-jB p105 and b-actin. Data are mean arbitrary units � SEM. White bars = placebo group;

black bars = ibuprofen group.

A B

Figure 3. NF-jB subunits p65 (A) and p50 (B) binding to nuclear protein. Data are mean arbitrary units � SEM. #denotes statistical significance

from 0 h post-exercise in the control group; $denotes statistical significance from 3 h post-exercise in the control group. White bars = placebo

group; black bars = ibuprofen group.

ª 2014 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf ofthe American Physiological Society and The Physiological Society.

2014 | Vol. 2 | Iss. 10 | e12172Page 7

L. Vella et al. NF-jB and Resistance Exercise

expression was elevated in the placebo group at 24 h

post-exercise. Coincident with this response, p50 tran-

scription factor binding activity was highest at 24 h

post-exercise. The biological significance of this delayed

increase in NF-jB p50 signalling during the latter stages

of post-exercise recovery remains uncertain. However, in

vitro work suggests that it may play a role in regulating

the active resolution of acute inflammation in skeletal

muscle (Lawrence et al. 2001, 2005; Senftleben et al.

2001). In a model of carageenin-induced pleurisy in

rats, Lawrence et al. (2001) provided preliminary evi-

dence for a complex interplay between distinct NF-jBsignalling pathways that control an acute and transient

inflammatory response. They reported that the prelimin-

ary phase of NF-jB activation that occurred at 6 h was

characteristic of the classical NF-jB pathway, and was

associated with the onset of acute inflammation. The

secondary phase was typical of the alternative NF-jBpathway, comprising p50 homodimers. Importantly,

inhibiting this wave of activity caused a prolonged

inflammatory response (Lawrence et al. 2001). Findings

from the present study support the concept of two

functionally distinct waves of NF-jB activity following

traditional resistance exercise. Future work is warranted

to determine how this secondary wave of NF-jB path-

way activity influences post-exercise inflammation and

skeletal muscle recovery.

We used ibuprofen supplementation to investigate

whether manipulating the inflammatory response to exer-

cise through the cyclooxygenase-prostaglandin pathway

alters post-exercise NF-jB signalling. Previous reports

indicate that NF-jB can function upstream of COX-2 to

control transcription of this gene (Pahl 1999; Poligone

and Baldwin 2001). Alternatively, prostaglandin activity

may also affect NF-jB (Rossi et al. 2000; Straus et al.

2000; Poligone and Baldwin 2001). In-vitro studies dem-

onstrate that the effect of prostaglandins on NF-jB activ-

ity is specific to the class of prostaglandin. Prostaglandin

E2 activates the classical NF-jB pathway, whereas prosta-

glandin A2, and prostaglandin J2 and its downstream

analogues can inhibit NF-jB activation in response to

pro-inflammatory stimuli (Castrillo et al. 2000; Rossi

et al. 2000; Straus et al. 2000; Lawrence et al. 2002). Our

group recently reported changes in prostaglandins in

blood serum samples after traditional resistance exercise

(Markworth et al. 2013). Interestingly, PGE2, PGA2 and

PGD2 all peaked in expression at 2 h post-exercise. We

did not find any significant time 9 group interaction

effects for the change in p65 phosphorylation; however,

LSD comparisons suggested that phosphorylated p65

expression only seemed to increase in the placebo group.

Similarly the observed changes in p50 protein expression

and DNA binding were only identified within the placebo

group. These findings are supported by data from the

study by Lawrence et al. (2001), which demonstrated no

change in the secondary wave of NF-jB DNA binding

activity following the administration of an alternative

COX inhibitor, NS398, in rats with pleurisy. While these

findings do not offer any conclusive evidence that ibupro-

fen treatment inhibits the NF-jB pathway, it does provide

justification to further explore this pathway as a potential

mechanism through which ibuprofen treatment inhibits

post-exercise inflammation.

There were several limitations to the present study that

need to be considered. Firstly, this analysis was run as

part of a larger study investigating the effects of ibuprofen

administration on multiple components of the post-exer-

cise inflammatory and hypertrophy response (Markworth

et al. 2013, 2014). Therefore, limited muscle sample

remained to complete further analyses. Future research

Table 2. Primer sequences were designed using Primer Express Software v 3.0 (Applied Biosystems) using sequences accessed through Gen-

bank and checked for specificity using nucleotide-nucleotide BLAST search.

Gene Accession No. Primer sequence

GAPDH NM_21130 Forward: CAT CCA TGA CAA CTT TGG TAT CGT

Reverse: CAG TCT TCT GGG TGG CAG TGA

MCP-1 NM_002982 Forward: TCC CAA AGA AGC TGT GAT CTT CA

Reverse: CAG ATT CTT GGG TGG AGT GA

IL-6 NM_000600 Forward: GCG AAA GGA TGA AAG TGA CCA T

Reverse: AGA CAA GCC CAG CAA TGA AAA

IL-8 NM_000584 Forward: CTG GCC GTC GCT CTC TGG

Reverse: TTA GCA CTC CTT GGC AAA ACT

TNF-a X_02910 Forward: GGA GAA GGG TGA CCG ACT CA

Reverse: TGC CCA GAC TCG GCA AAG

COX-2 U_20548 Forward: GAA TCA TTC ACC AGG CAA ATT G

Reverse: TGG AAG CCT GTG ATA CTT TCT GTA CT

2014 | Vol. 2 | Iss. 10 | e12172Page 8

ª 2014 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of

the American Physiological Society and The Physiological Society.

NF-jB and Resistance Exercise L. Vella et al.

should consider the post-exercise regulation of upstream

markers including IKKa and IKKb.

Conclusion

This research provides new insights into the regulation of

NF-jB following a bout of acute resistance exercise. The

primary finding from this study is that NF-jB activation

follows a biphasic activation pattern that is subunit-spe-

cific. The first wave involves the classical NF-jB p65 sub-

unit, and corresponds with the onset of an acute

inflammatory response and an increase in inflammatory

cytokine gene expression. The second wave is consistent

with a previously identified secondary wave of NF-jB

activity that involves the alternative p50 subunit. Research

in alternative models of inflammation suggests that this

second wave of activity may be associated with an active

inflammatory resolution program. Further research into

the role of p50 signalling in skeletal muscle represents a

key area for future research in order to better understand

the mechanisms that regulate post-exercise inflammation.

Analysis of the LSD to examine pair-wise within-group

differences suggested that the observed changes in both

NF-jB p65 and p50 were detected only within the pla-

cebo group. The complex interplay between the NF-jBand the COX/prostaglandin pathway in exercise-induced

muscle damage remains poorly understood, and should

be a focus for future research.

A B

C D

E

Figure 4. RT-PCR analysis of NF-jB target genes IL-6 (A), IL-8 (B), MCP-1 (C), TNF-a (D) and COX-2 (E) in skeletal muscle cDNA. Data are mean

arbitrary units � SEM. *denotes statistical significance from pre exercise in the same treatment group; #denotes statistical significance from 0 h

post-exercise in the same treatment group; ^denotes statistical significance from 24 h post-exercise in the same treatment group (P < 0.05).

White bars = placebo group; black bars = ibuprofen group.

ª 2014 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf ofthe American Physiological Society and The Physiological Society.

2014 | Vol. 2 | Iss. 10 | e12172Page 9

L. Vella et al. NF-jB and Resistance Exercise

Acknowledgments

The authors would like to acknowledge to assistance of

Associate Professor John Reynolds for his contributions

in the statistical analysis of the dataset.

Conflict of Interest

There were no conflicts of interest relevant to this paper.

References

Bakkar, N., and D. C. Guttridge. 2010. NF-kappaB signaling: a

tale of two pathways in skeletal myogenesis. Physiol. Rev.

90:495–511.

Bohuslav, J., V. V. Kravchenko, G. C. Parry, J. H. Erlich,

S. Gerondakis, N. Mackman, et al. 1998. Regulation of an

essential innate immune response by the p50 subunit of

NF-kappaB. J. Clin. Investig. 102:1645–1652.

Bonizzi, G., and M. Karin. 2004. The two NF-kappaB

activation pathways and their role in innate and adaptive

immunity. Trends Immunol. 25:280–288.

Buford, T. W., M. B. Cooke, and D. S. Willoughby. 2009.

Resistance exercise-induced changes of inflammatory gene

expression within human skeletal muscle. Eur. J. Appl.

Physiol. 107:463–471.

Castrillo, A., M. J. D�ıaz-Guerra, S. Hortelano, P. Mart�ın-Sanz,

and L. Bosc�a. 2000. Inhibition of IkappaB kinase and

IkappaB phosphorylation by 15-deoxy-Delta

(12,14)-prostaglandin J(2) in activated murine macrophages.

Mol. Cell. Biol. 20:1692–1698.

Delhalle, S., R. Blasius, M. Dicato, and M. Diederich. 2004.

A beginner’s guide to NF-kappaB signaling pathways. Ann.

N. Y. Acad. Sci. 1030:1–13.

Dodd, S. L., B. Hain, S. M. Senf, and A. R. Judge. 2009.

Hsp27 inhibits IKKb-induced NF-jB activity and skeletal

muscle atrophy. FASEB J. 23:3415–3423.

Durham, W. J., L. Yi-Ping, E. Gerken, M. Farid, S. Arbogast,

R. R. Wolfe, et al. 2004. Fatiguing exercise reduces DNA

binding activity of NF-jB in skeletal muscle nuclei. J. Appl.

Physiol. 97:1740–1745.

Ho, R. C., M. F. Hirshman, Y. Li, D. Cai, J. R. Farmer,

W. G. Aschenbach, et al. 2005. Regulation of IkappaB kinase

and NF-kappaB in contracting adult rat skeletal muscle.

Am. J. Physiol. Cell Physiol. 289:C794–C801.

Hohmann, H. P., R. Remy, C. Scheidereit, and A. P. van

Loon. 1991. Maintenance of NF-kappa B activity is

dependent on protein synthesis and the continuous presence

of external stimuli. Mol. Cell. Biol. 11:259–266.

Hollander, J., R. Fiebig, M. Gore, T. Ookawara, H. Ohno, and

L. L. Ji. 2001. Superoxide dismutase gene expression is

activated by a single bout of exercise in rat skeletal muscle.

Pfl€ugers Arch. 442:426–434.

Hyldahl, R. D., L. Xin, M. J. Hubal, S. Moeckel-Cole,

S. Chipkin, and P. M. Clarkson. 2011. Activation of nuclear

factor-jB following muscle eccentric contractions in humans

is localized primarily to skeletal muscle-residing pericytes.

FASEB J. 25:2956–2966.

Ishikawa, H., E. Claudio, D. Dambach, C. Ravent�os-Su�arez,

C. Ryan, and R. Bravo. 1998. Chronic inflammation and

susceptibility to bacterial infections in mice lacking the

polypeptide (p)105 precursor (NF-kappaB1) but expressing

p50. J. Exp. Med. 187:985–996.

Ji, L. L., M. C. Gomez-Cabrera, N. Steinhafel, and J. Vina.

2004. Acute exercise activates nuclear factor (NF)-kappaB

signaling pathway in rat skeletal muscle. FASEB J. 18:

1499–1506.

Kramer, H. F., and L. J. Goodyear. 2007. Exercise, MAPK, and

NF-kappaB signaling in skeletal muscle. J. Appl. Physiol.

103:388–395.

Lawrence, T., D. W. Gilroy, P. R. Colville-Nash, and

D. A. Willoughby. 2001. Possible new role for NF-kappaB in

the resolution of inflammation. Nat. Med. 7:1291–1297.

Lawrence, T., D. A. Willoughby, and D. W. Gilroy. 2002.

Anti-inflammatory lipid mediators and insights into the

resolution of inflammation. Nat. Rev. Immunol. 2:787–795.

Lawrence, T., M. Bebien, G. Y. Liu, V. Nizet, and M. Karin.

2005. IKKalpha limits macrophage NF-kappaB activation

and contributes to the resolution of inflammation. Nature

434:1138–1143.

Malm, C., P. Nyberg, M. Engstrom, B. Sjodin, R. Lenkei,

B. Ekblom, et al. 2000. Immunological changes in human

skeletal muscle and blood after eccentric exercise and

multiple biopsies. J. Physiol. 529(Pt 1):243–262.

Markworth, J. F., L. Vella, B. S. Lingard, D. L. Tull,

T. W. Rupasinghe, A. J. Sinclair, et al. 2013. Human

inflammatory and resolving lipid mediator responses to

resistance exercise and ibuprofen treatment. Am. J. Physiol.

Regul. Integr. Comp. Physiol. 305:R1281–R1296.

Markworth, J. F., L. D. Vella, V. C. Figueiredo, and

D. Cameron-Smith. 2014. Ibuprofen treatment blunts early

translational signaling responses in human skeletal muscle

following resistance exercise. J. Appl. Physiol. 117:20–28.

Mayhew, J. L., J. L. Prinster, J. S. Ware, D. L. Zimmer,

J. R. Arabas, and M. G. Bemben. 1995. Muscular endurance

repetitions to predict bench press strength in men of

different training levels. J. Sports Med. Phys. Fitness 35:

108–113.

Mead, R.. 1988. The design of experiments: statistical

principles for practical applications. Cambridge University

Press, New York, NY.

Mikkelsen, U. R., H. Langberg, I. C. Helmark,

D. Skovgaard, L. L. Andersen, M. Kjaer, et al. 2009. Local

NSAID infusion inhibits satellite cell proliferation in

human skeletal muscle after eccentric exercise. J. Appl.

Physiol. 107:1600–1611.

Mourkioti, F., and N. Rosenthal. 2008. NF-jB signaling in

skeletal muscle: prospects for intervention in muscle

diseases. J. Mol. Med. 86:747–759.

2014 | Vol. 2 | Iss. 10 | e12172Page 10

ª 2014 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of

the American Physiological Society and The Physiological Society.

NF-jB and Resistance Exercise L. Vella et al.

Pahl, H. L. 1999. Activators and target genes of Rel/

NF-kappaB transcription factors. Oncogene 18:6853–6866.

Paulsen, G., U. R. Mikkelsen, T. Raastad, and J. M. Peake.

2012. Leucocytes, cytokines and satellite cells: what role do

they play in muscle damage and regeneration following

eccentric exercise? Exerc. Immunol. Rev. 18:42–97.

Poligone, B., and A. S. Baldwin. 2001. Positive and negative

regulation of NF-kappaB by COX-2: roles of different

prostaglandins. J. Biol. Chem. 276:38658–38664.

Rossi, A., P. Kapahi, G. Natoli, T. Takahashi, Y. Chen,

M. Karin, et al. 2000. Anti-inflammatory cyclopentenone

prostaglandins are direct inhibitors of IkappaB kinase.

Nature 403:103–108.

Saville, D. J. 1990. Multiple comparison procedures: the

practical solution. Am. Stat. 44:174.

Senftleben, U., Y. Cao, G. Xiao, F. R. Greten, G. Kr€ahn,

G. Bonizzi, et al. 2001. Activation by IKKalpha of a second,

evolutionary conserved, NF-kappa B signaling pathway.

Science 293:1495–1499.

Spangenburg, E. E., D. A. Brown, M. S. Johnson, and

R. L. Moore. 2006. Exercise increases SOCS-3 expression in

rat skeletal muscle: potential relationship to IL-6 expression.

J. Physiol. 572:839–848.

Straus, D. S., G. Pascual, M. Li, J. S. Welch, M. Ricote,

C. H. Hsiang, et al. 2000. 15-deoxy-delta

12,14-prostaglandin J2 inhibits multiple steps in the

NF-kappa B signaling pathway. Proc. Natl Acad. Sci. USA

97:4844–4849.

Tantiwong, P., K. Shanmugasundaram, A. Monroy, S. Ghosh,

M. Li, R. A. DeFronzo, et al. 2010. NF-jB activity in muscle

from obese and type 2 diabetic subjects under basal and

exercise-stimulated conditions. Am. J. Physiol. Endocrinol.

Metab. 299:E794–E801.

Trappe, T. A., F. White, C. P. Lambert, D. Cesar,

M. Hellerstein, and W. J. Evans. 2002. Effect of ibuprofen

and acetaminophen on postexercise muscle protein

synthesis. Am. J. Physiol. Endocrinol. Metab. 45:E551.

Trenerry, M. K., K. A. Carey, A. C. Ward, and

D. Cameron-Smith. 2007. STAT3 signaling is activated in

human skeletal muscle following acute resistance exercise.

J. Appl. Physiol. 102:1483–1489.

Urso, M. L.. 2013. Anti-inflammatory interventions and

skeletal muscle injury: benefit or detriment? J. Appl. Physiol.

115: 920–928.

Vella, L., M. K. Caldow, A. E. Larsen, D. Tassoni, P. A. Della

Gatta, P. Gran, et al. 2012. Resistance exercise increases

NF-jB activity in human skeletal muscle. Am. J. Physiol.

Regul. Integr. Comp. Physiol. 302:R667–R673.

Whisenant, M. J., L. B. Panton, W. B. East, and

C. E. Broeder. 2003. Validation of submaximal prediction

equations for the 1 repetition maximum bench press test on

a group of collegiate football players. J. Strength Cond. Res.

17:221–227.

Xin, L., R. D. Hyldahl, S. R. Chipkin, and P. M. Clarkson.

2013. A contralateral repeated bout effect attenuates

induction of NF-?B DNA-binding following eccentric

exercise. J. Appl. Physiol. 116:1473–1478.

Zandi, E., D. M. Rothwarf, M. Delhase, M. Hayakawa, and

M. Karin. 1997. The IkappaB kinase complex (IKK)

contains two kinase subunits, IKKalpha and IKKbeta,

necessary for IkappaB phosphorylation and NF-kappaB

activation. Cell 91:243–252.

ª 2014 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf ofthe American Physiological Society and The Physiological Society.

2014 | Vol. 2 | Iss. 10 | e12172Page 11

L. Vella et al. NF-jB and Resistance Exercise

145

Appendix C:

146

147