26.07.2013 Views

Identification of important interactions between subchondral bone ...

Identification of important interactions between subchondral bone ...

Identification of important interactions between subchondral bone ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

F A C U L T Y O F S C I E N C E<br />

U N I V E R S I T Y O F C O P E N H A G E N<br />

<strong>Identification</strong> <strong>of</strong> <strong>important</strong> <strong>interactions</strong><br />

<strong>between</strong> <strong>subchondral</strong> <strong>bone</strong> turnover and<br />

cartilage degradation in the pathogenesis <strong>of</strong><br />

osteoarthritis<br />

PhD thesis by Suzi Høgh Madsen M.Sc<br />

November 2011


Front page pictures<br />

Digital drawing <strong>of</strong> a knee symbolizing pain (http://www.spa-resorts.cz/eng/kaleidoskop/osteoarthritis-treatment-1196.html).<br />

Logo from the sponsor <strong>of</strong> my industrial PhD; The ministry <strong>of</strong> Science, Technology and Innovation.<br />

Logo from the company where I performed my industrial PhD; Nordic Bioscience A/S.<br />

Drawing <strong>of</strong> the three cell types investigated in the present thesis; Osteoclast (red), osteoblast (brawn), and<br />

chondrocyte (blue).


Supervisors<br />

University <strong>of</strong> Copenhagen:<br />

Supervisors<br />

Søren Tvorup Christensen, PhD, Associate Pr<strong>of</strong>essor<br />

Department <strong>of</strong> Biology<br />

Universitetsparken 13<br />

DK-2100 Copenhagen OE<br />

stchristensen@bio.ku.dk<br />

Nordic Bioscience A/S:<br />

Morten Asser Karsdal, PhD, CEO<br />

Herlev Hovedgade 207<br />

DK-2730 Herlev<br />

mk@nordicbioscience.com<br />

Anne-Christine Bay-Jensen, PhD<br />

Herlev Hovedgade 207<br />

DK-2730 Herlev<br />

acbj@nordicbioscience.com<br />

Kim Henriksen, PhD<br />

Herlev Hovedgade 207<br />

DK-2730 Herlev<br />

kh@nordicbioscience.com<br />

1


Acknowledgements<br />

Acknowledgements<br />

First, I would like to acknowledge the Ministry <strong>of</strong> Science, Technology and Innovation (VTU)<br />

and Nordic Bioscience A/S, who funded this industrial Ph.D project.<br />

I would like to thank my supervisors Morten Asser Karsdal from Nordic<br />

Bioscience and Søren Tvorup Christensen from the University <strong>of</strong> Copenhagen, who have both<br />

provided great scientific guidance and enthusiasm throughout the project and writing process. I<br />

would also like to thank the co-supervisors, Anne-Christine Bay-Jensen and Kim Henriksen, for<br />

their support, excellent coaching, and helpful scientific discussions throughout my time at Nordic<br />

Bioscience. Furthermore, I would like to thank all my colleagues at Nordic Bioscience for helpful<br />

comments, technical help, scientific discussions, good times, and for creating a warm and<br />

pleasant work environment.<br />

Finally, I would like to thank my family and friends who have always supported my<br />

dreams and ambitions. Especially, I would like to thank Rasmus Klinck, for critical pro<strong>of</strong>reading<br />

<strong>of</strong> my thesis, as well as for good friendship and support throughout my thesis.<br />

__________________________________<br />

Suzi Høgh Madsen<br />

November 2011<br />

2


Preface<br />

Preface<br />

The work presented in this thesis was performed in collaboration <strong>between</strong> Nordic Bioscience<br />

A/S and the University <strong>of</strong> Copenhagen under the industrial PhD agreement and with the<br />

supervision <strong>of</strong> Pr<strong>of</strong>. Morten Asser Karsdal (company supervisor), Dr. Anne-Christine Bay Jensen<br />

(project supervisor, company), Dr. Kim Henriksen (project supervisor, company), and Associate<br />

Pr<strong>of</strong>. Søren Tvorup Christensen (university supervisor). The work was carried out in the period<br />

ranging from October 2008 to November 2011.<br />

The PhD work includes four original research papers listed below. Three papers<br />

have been accepted and published, and one is under review. The individual articles are referred to<br />

in the present thesis by their roman numerals.<br />

The PhD thesis consists <strong>of</strong> an introduction to the topics <strong>of</strong> the four papers, the<br />

four papers, and a general conclusion.<br />

Paper I:<br />

Characterization <strong>of</strong> an Ex vivo Femur Head Model Assessed by Markers <strong>of</strong> Bone and<br />

Cartilage Turnover<br />

Suzi Hoegh Madsen, Anne S<strong>of</strong>ie Goettrup, Gedske Thomsen, Søren Tvorup Christensen,<br />

Nikolaj Schultz, Kim Henriksen, Anne-Christine Bay-Jensen, and Morten Asser Karsdal<br />

Cartilage 2011;2(3):265–278<br />

Paper II:<br />

Glucocorticoids exert context-dependent effects on cells <strong>of</strong> the joint in vitro<br />

Suzi H. Madsen, Kim V. Andreassen, Søren T. Christensen, Morten A. Karsdal,<br />

Francis M. Sverdrup, Anne-Christine Bay-Jensen, and Kim Henriksen<br />

Steroids 2011;76(13):1474-1482<br />

Paper III:<br />

Aggrecanase- and matrix metalloproteinase-mediated aggrecan degradation is associated<br />

with different molecular characteristics <strong>of</strong> aggrecan and separated in time ex vivo<br />

Suzi Hoegh Madsen, Eren Ufuk Sumer, Anne-Christine Bay-Jensen, Bodil-Cecilie Sondergaard,<br />

Per Qvist, and Morten Asser Karsdal<br />

Biomarkers 2010;15(3):266-276<br />

Paper IV:<br />

Metalloproteinase and cysteine protease pr<strong>of</strong>iling in normal and Osteoarthritic cartilage<br />

Suzi Hoegh Madsen, Kim Henriksen, Søren Tvorup Christensen, Morten Asser Karsdal, and<br />

Anne Christine Bay-Jensen<br />

In review in Proteome Science<br />

The industrial PhD project has been supported by the Ministry <strong>of</strong> Science, Technology and<br />

Innovation (VTU) (08-036109) and Nordic Bioscience A/S.<br />

3


Suzi Høgh Madsen, PhD thesis<br />

4


Contents<br />

Contents<br />

Abstract .......................................................................................................................................................... 7<br />

Sammendrag på dansk (abstract in Danish) ............................................................................................. 9<br />

CHAPTER 1<br />

Objectives .................................................................................................................................................... 11<br />

CHAPTER 2<br />

Introduction ................................................................................................................................................ 13<br />

2.1 Epidemiology and etiology <strong>of</strong> osteoarthritis ................................................................................ 13<br />

2.2 Biology <strong>of</strong> the joint .......................................................................................................................... 15<br />

2.2.1 Development <strong>of</strong> <strong>bone</strong> from a cartilage template ................................................................. 15<br />

2.2.2 Adult <strong>bone</strong> - macroscopic and microscopic organization .................................................. 17<br />

2.2.3 The cells <strong>of</strong> <strong>bone</strong> and their communications ....................................................................... 19<br />

2.2.4 Articular cartilage ...................................................................................................................... 22<br />

2.3 The pathology <strong>of</strong> Osteoarthritis .................................................................................................... 25<br />

2.3.1 A chondrocyte is not just a chondrocyte .............................................................................. 26<br />

2.3.2 The extracellular matrix <strong>of</strong> cartilage in OA .......................................................................... 28<br />

2.3.3 The <strong>subchondral</strong> <strong>bone</strong> in OA ................................................................................................. 29<br />

2.3.4 Diagnostics - biochemical markers as a tool ........................................................................ 30<br />

2.4 The effect <strong>of</strong> Glucocorticoids ........................................................................................................ 33<br />

2.5 References for CHAPTER 2 ......................................................................................................... 35<br />

CHAPTER 3<br />

Overview <strong>of</strong> OA models ........................................................................................................................... 43<br />

3.1 Pre-clinical models ........................................................................................................................... 43<br />

3.2 Advantages/disadvantages <strong>of</strong> pre-clinical models ...................................................................... 44<br />

3.2.1 In vitro .......................................................................................................................................... 44<br />

3.2.2 Ex vivo......................................................................................................................................... 44<br />

3.2.3 In vivo........................................................................................................................................... 46<br />

3.3 Ex vivo controls ................................................................................................................................ 47<br />

3.4 References for CHAPTER 3 ......................................................................................................... 48<br />

5


Contents<br />

CHAPTER 4<br />

PAPER I ...................................................................................................................................................... 51<br />

CHAPTER 5<br />

PAPER II .................................................................................................................................................... 67<br />

CHAPTER 6<br />

PAPER III ................................................................................................................................................... 77<br />

CHAPTER 7<br />

PAPER IV ................................................................................................................................................... 89<br />

CHAPTER 8<br />

General discussion and conclusions ...................................................................................................... 101<br />

Future perspectives .................................................................................................................................. 103<br />

Abbreviations ............................................................................................................................................ 104<br />

Appendix I ................................................................................................................................................. 105<br />

Co-author declarations for paper I - IV .......................................................................................... 105<br />

Appendix II ............................................................................................................................................... 112<br />

Publication list ...................................................................................................................................... 112<br />

6


Abstract<br />

Abstract<br />

Osteoarthritis (OA) is the most common form <strong>of</strong> arthritis and a major cause <strong>of</strong> pain and<br />

disability, with prevalence increasing with age. At present there are no structure modifying<br />

treatments accepted by the Food and Drug Administration (FDA), emphasizing the need for<br />

understanding the pathogenic processes leading to OA. OA is a complex disease <strong>of</strong> the entire<br />

joint, including <strong>bone</strong>, cartilage and the synovium, and it is characterized by the progressive<br />

degradation <strong>of</strong> articular cartilage, mild synovial inflammation, and alterations <strong>of</strong> the <strong>subchondral</strong><br />

<strong>bone</strong>. Currently, it is not clear whether the pathogenesis <strong>of</strong> OA originates in the <strong>bone</strong> or cartilage<br />

compartment, and little is still known on how these compartments drive disease progression. An<br />

increased amount <strong>of</strong> evidence suggests a strong coupling <strong>between</strong> the <strong>subchondral</strong> <strong>bone</strong> and the<br />

articular cartilage turnover, with pathological processes occurring simultaneously in both<br />

compartments. Therefore, an optimal intervention strategy for OA likely includes targeting both<br />

<strong>bone</strong> and cartilage compartments.<br />

The aim <strong>of</strong> the thesis was to investigate the pathogenesis <strong>of</strong> OA with respect to<br />

<strong>bone</strong> and cartilage, and the importance <strong>of</strong> the interaction <strong>between</strong> the two tissues. Understanding<br />

the <strong>interactions</strong> <strong>between</strong> osteoclasts, osteoblast and chondrocytes may be essential for the<br />

development <strong>of</strong> future treatments for OA.<br />

Before the launch <strong>of</strong> this thesis, there were no validated pre-clinical models that<br />

allowed investigation <strong>of</strong> whole tissue pathology <strong>of</strong> OA in one single system, which combined the<br />

metabolism <strong>of</strong> the implicated cell types. Thus, the present thesis focused on the development and<br />

characterization <strong>of</strong> a novel murine ex vivo femur head model, comprising the three major cell<br />

types involved in the deterioration <strong>of</strong> joint structure; the osteoblasts, osteoclasts, and<br />

chondrocytes. We have established the ex vivo femur head model with positive and negative<br />

controls, which allow the investigation <strong>of</strong> the interaction <strong>between</strong> <strong>bone</strong> and cartilage.<br />

Furthermore, by using the novel ex vivo model and other pre-clinical models, we evaluated the<br />

effect <strong>of</strong> glucocorticoids on <strong>bone</strong> and cartilage and found that the effect <strong>of</strong> glucocorticoids highly<br />

depend on the activation and differential stage <strong>of</strong> the cell targeted in the joint.<br />

The thesis also studied the degradation processes <strong>of</strong> cartilage, focusing on the two<br />

predominant proteins; aggrecan and collagen type II. We investigated the molecular differences<br />

<strong>between</strong> matrix metalloproteinase- and aggrecanase-mediated aggrecan degradation as a<br />

consequence <strong>of</strong> their distinct time-dependent degradation pr<strong>of</strong>iles. We found that the aggrecan<br />

molecule may be divided into two different protease-specific pools. Furthermore, we investigated<br />

the differences <strong>between</strong> normal and OA cartilage with respect to endogenous proteases (assessed<br />

by biochemical markers) and found that the degradation markers are released in part by different<br />

proteolytic pathways.<br />

7


Abstract<br />

In conclusion, we have established a pre-clinical whole-tissue model for investigating cell-cell<br />

<strong>interactions</strong> that reflect parts <strong>of</strong> the processes in the pathogenesis <strong>of</strong> joint degenerative diseases.<br />

This model is also an excellent screening model for potential OA drugs. The model confirmed<br />

beneficial effects <strong>of</strong> glucocorticoids previously observed on articular cartilage, but not <strong>bone</strong>. The<br />

loss <strong>of</strong> sulphated glycosaminoglycans (sGAGs) from cartilage in early OA is not equivalent to<br />

loss <strong>of</strong> aggrecan, since there may be different pools <strong>of</strong> aggrecan that are sensitive to different<br />

types <strong>of</strong> proteases. Furthermore, the role <strong>of</strong> endogenous proteases probably depends on the stage<br />

<strong>of</strong> OA, as we did not observe the same release <strong>of</strong> biochemical markers in normal versus OA<br />

cartilage. This highlights the importance <strong>of</strong> developing different treatments specific for the<br />

different stages <strong>of</strong> OA.<br />

8


Sammendrag på dansk<br />

Sammendrag på dansk (abstract in Danish)<br />

Slidgigt er den mest almindelige form for gigt der findes og en væsentlig årsag til smerte og<br />

invalidering. Risikoen for at få slidgigt øges betydeligt med alderen. På nuværende tidspunkt<br />

findes der ingen godkendte behandlinger for slidgigt fra den amerikanske Food and Drug<br />

Administration (FDA), hvilket understreger behovet for at forstå de patogene processer, der<br />

fører til sygdommen. Slidgigt er en kompleks sygdom, der involverer hele leddet, hvilket<br />

inkluderer bl.a. knogle, brusk og synovium. Slidgigt kendetegnes ved en gradvis nedbrydning af<br />

ledbrusken, en mild inflammation af synovial-vævet og ændringer af den <strong>subchondral</strong>e knogle. I<br />

øjeblikket diskuteres det om patogenesen af slidgigt starter i knogle- eller brusk-vævet, og om<br />

vævene hver især er med til at gøre sygdomsforløbet værre. En del forskning tyder på at der i<br />

slidgigt er sammenhæng mellem ændringer i <strong>subchondral</strong> knogle, ændringer i ledbrusken, og de<br />

patogene processer der forekommer samtidigt i begge væv. Derfor vil den bedste behandling af<br />

slidgigt sandsynligvis være en behandling der både involverer knogler og brusk.<br />

Formålet med PhD-afhandlingen var at undersøge patogenesen af slidgigt med<br />

fokus på knogle- og brusk-delen, og at undersøge hvor vigtig interaktionen er mellem de to væv.<br />

Det kan være afgørende for udviklingen af fremtidige behandlinger for slidgigt at forstå det<br />

samspil, der er mellem osteoklaster, osteoblaster og kondrocytter.<br />

Før jeg startede på denne afhandling, var der ingen etablerede prækliniske modeller,<br />

der kunne undersøge de patologiske processer, der forekommer i slidgigt i ét enkelt system, og<br />

som indeholder de implicerede celletyper fra leddet. Derfor fokuserede denne afhandling på<br />

udviklingen og karakteriseringen af en ny ex vivo femur hoved muse-model, der indeholder de tre<br />

hoved-celletyper, som er involveret i degenereringen af leddet; osteoblaster, osteoklaster og<br />

kondrocytter. Vi har etableret en ny ex vivo femur hoved model med positive og negative<br />

kontroller, der kan undersøge interaktionen mellem knogle og brusk. Herudover, ved at bruge<br />

den nye ex vivo model og andre prækliniske modeller, undersøgte vi effekten af glukokortikoider<br />

på knogle og brusk, og fandt at effekten af glukokortikoider afhænger betydeligt af aktiveringen<br />

og differentieringsstadiet af den undersøgte celle fra leddet.<br />

Afhandlingen undersøgte også de nedbrydningsprocesser vi finder i brusk, med<br />

fokus på de to hoved-proteiner; aggrecan og kollagen type II. Vi undersøgte de molekylære<br />

forskelle mellem matrix metalloproteinase- og aggrecanase-genereret aggrecan nedbrydning, på<br />

grund af deres forskellige nedbrydningspr<strong>of</strong>iler over tid. Vi fandt at aggrecanmolekylet måske er<br />

opdelt i to forskellige proteasespecifikke puljer. Desuden undersøgte vi de forskelle, der er<br />

mellem normalt og sygt brusk mht. endogene proteaser (målt med biokemiske markører) og<br />

fandt at nedbrydnings-markørerne til dels er frigivet via forskellige proteolytiske veje.<br />

9


Sammendrag på dansk<br />

Samlet set har vi etableret en præklinisk fler-vævs model, der kan undersøge celle-celle<br />

interaktioner, og som afspejler dele af processerne, man ser i patogenesen af degenerative led-<br />

sygdomme. Denne model er også en udmærket screeningsmodel for potentielle medikamenter<br />

for slidgigt. Modellen bekræftede de gavnlige virkninger af glukokortikoider man tidligere har set<br />

på ledbrusk, men ikke på knogle. Udtømning af sulfat-glykosaminoglycaner (sGAG’s) fra brusk i<br />

begyndende slidgigt er ikke ensbetydende med udtømning af aggrecaner, da vi foreslår, at der er<br />

forskellige puljer af aggrecaner, der er følsomme over for forskellige typer af proteaser.<br />

Derudover afhænger rollerne for de endogene proteaser formentlig af det slidgigt-stadie man er i,<br />

da vi fandt forskellige frigivelsesmønstre af biokemiske markører i normalt brusk versus slidgigt<br />

brusk. Dette understreger betydningen af at udvikle forskellige behandlinger, der er specifikke for<br />

de forskellige stadier af slidgigt.<br />

10


CHAPTER 1<br />

Objectives<br />

CHAPTER 1: Objectives<br />

The aim <strong>of</strong> the PhD project was to gain further insight into the pathogenesis <strong>of</strong> osteoarthritis<br />

(OA) with respect to <strong>bone</strong> and cartilage, which are believed to be the primary tissues involved in<br />

joint turnover. In particular, the importance <strong>of</strong> the interaction <strong>between</strong> <strong>bone</strong> and cartilage was<br />

investigated to assess the importance <strong>of</strong> <strong>bone</strong> in the pathogenesis <strong>of</strong> OA. To this end, we wanted<br />

to develop a pre-clinical model that includes the two key tissues, <strong>subchondral</strong> <strong>bone</strong> and articular<br />

cartilage. Furthermore, as articular cartilage degradation is believed to be a hallmark <strong>of</strong> OA<br />

pathogenesis, we investigated the proteolytic processes <strong>of</strong> pathogenic cartilage in order to<br />

understand the OA-induced changes, which ultimately lead to cartilage loss.<br />

The PhD project had two main objectives:<br />

1) The commercial objective.<br />

The first objective was to develop and validate a pre-clinical model system that allows<br />

investigation <strong>of</strong> whole tissue pathology in the pathogenesis <strong>of</strong> OA. This model should contain<br />

the three major cell types involved in the deterioration <strong>of</strong> joint structure, the osteoblasts,<br />

osteoclasts, and chondrocytes, including their cellular signalling processes (PAPER I). This model<br />

should allow testing <strong>of</strong> potential OA drugs in a more pathological relevant model. Thus, the<br />

effects <strong>of</strong> glucocorticoids as a potential drug for OA were tested in this model to evaluate the<br />

usefulness <strong>of</strong> the model and further to evaluate the effect <strong>of</strong> glucocorticoids on <strong>bone</strong> and<br />

cartilage cells (PAPER II).<br />

2) The basic research objective.<br />

The second objective focused on the degradation <strong>of</strong> collagen type II and aggrecan in OA<br />

cartilage. We hypothesized that the loss <strong>of</strong> aggrecan in early and late OA was dependent on<br />

different proteases due to different molecular structures <strong>of</strong> the aggrecan. We expected to find<br />

different pools <strong>of</strong> aggrecan being degraded by appertaining proteases (so a specific protease<br />

should be targeted depending on the disease stage) (PAPER III). We also hypothesized that the<br />

ratio <strong>between</strong> endogenous proteases in OA cartilage, depends on the stage <strong>of</strong> the disease.<br />

Furthermore, we wanted to investigate if the biochemical markers were able to assess the<br />

different endogenous proteases in normal vs. OA cartilage (PAPER IV).<br />

11


Suzi Høgh Madsen, PhD thesis<br />

12


CHAPTER 2<br />

Introduction<br />

CHAPTER 2: Introduction<br />

This Introduction aims to introduce the knowledge that forms the basis for my thesis work. The<br />

scope <strong>of</strong> the introduction is: i) to provide an overview <strong>of</strong> the development and biology <strong>of</strong> the<br />

joint, ii) the degeneration <strong>of</strong> the joint, with emphasis on osteoarthritis (OA), iii) and to introduce<br />

the current knowledge on the mode <strong>of</strong> action <strong>of</strong> glucocorticoids.<br />

2.1 Epidemiology and etiology <strong>of</strong> osteoarthritis<br />

There are more than 100 different types <strong>of</strong> arthritis 1 . The most common type is OA, also known<br />

as degenerative joint disease or degenerative arthritis. It is a condition that affects more than 46<br />

million people in the USA, and it is expected to affect 67 million people by the year 2030 1 .<br />

According to Gigtforeningen (The Danish Arthritis Society), more than 210.000 people in Denmark<br />

are diagnosed with OA, but experts estimate that the actual number is much higher. Although the<br />

disease occurs in people <strong>of</strong> all ages, it is most common in the older population; 50% <strong>of</strong> people<br />

over the age <strong>of</strong> 40 years and 100% <strong>of</strong> people over the age <strong>of</strong> 60 years have OA in one or more<br />

joints 2 . Epidemiologic studies further suggest that there are clear sex-specific differences 3 . Before<br />

50 years <strong>of</strong> age, the prevalence <strong>of</strong> OA in most joints is higher in men than in women. After the<br />

age <strong>of</strong> 50 years, women are more <strong>of</strong>ten affected with hand-, foot-, and knee OA than men 4 .<br />

The etiology <strong>of</strong> OA is unknown. The general perception <strong>of</strong> OA is that it is a<br />

cartilage disease. However, OA is not only affecting the articular cartilage, but the entire joint,<br />

including the <strong>subchondral</strong> <strong>bone</strong>, ligaments, joint capsule, synovial membrane and periarticular<br />

muscles. However, loss <strong>of</strong> cartilage is the obvious central hallmark <strong>of</strong> OA 5 . An increasing line <strong>of</strong><br />

evidence suggests that both <strong>bone</strong> and cartilage contribute to the onset <strong>of</strong> the disease. One <strong>of</strong> the<br />

first indications came from clinical observations suggesting that osteopenic women generally do<br />

not develop severe OA 6 . Likewise, subjects with OA <strong>of</strong> the hip have greater <strong>bone</strong> mass than<br />

normal subjects or subjects with osteoporosis 7 , suggesting that OA could initially be a disease <strong>of</strong><br />

the <strong>bone</strong> 8 . To date, the majority <strong>of</strong> clinical studies support the idea that OA is associated with<br />

increased <strong>bone</strong> density, but the subject remains controversial 9 .<br />

OA commonly affects hands, feet, spine, and large weight-bearing joints, such as<br />

the hip and knees. In most cases, the mechanisms underlying the development <strong>of</strong> OA are<br />

unknown. These cases are classified as primary OA and are mostly related to aging or heredity.<br />

When the cause <strong>of</strong> OA is known, as after an injury, trauma, anatomic abnormalities or obesity,<br />

the condition is referred to as secondary OA 5,10,11 . OA is characterized by gradual loss <strong>of</strong> articular<br />

cartilage, alterations <strong>of</strong> the <strong>subchondral</strong> <strong>bone</strong>, and development <strong>of</strong> osteophytes. These alterations<br />

13


CHAPTER 2: Introduction<br />

lead to pain and limitation <strong>of</strong> joint mobility, which decrease the quality <strong>of</strong> life for the patients and<br />

become a burden on the society 5 . Today, treatment <strong>of</strong> OA is based on anti-inflammatory drugs<br />

(e.g. NSAID), analgesics or arthroplastic surgery 12 , which neither slow down nor cure the disease.<br />

Moreover, no disease modifying osteoarthritic drugs (DMOADS) have been developed and<br />

approved by the American Food and Drug Administration (FDA) yet. Therefore, the discovery<br />

and the development <strong>of</strong> effective disease-modifying drugs are urgently needed.<br />

The large number <strong>of</strong> people affected by OA is increasing<br />

very fast. The increased life expectancy <strong>of</strong> the elderly<br />

population and the obesity epidemic, affirms the necessity<br />

to find a treatment for OA as fast a possible: Both to<br />

prevent an “OA-epidemic”, but also to avoid the great<br />

social-economical costs in the future.<br />

14


2.2 Biology <strong>of</strong> the joint<br />

CHAPTER 2: Introduction<br />

In order to get insight into the underlying processes <strong>of</strong> OA, it is <strong>important</strong> to understand the<br />

physiology and biology <strong>of</strong> healthy joints. Joints that are able to move freely are termed synovial<br />

joints and together with muscles and <strong>bone</strong> they represent the machinery that enables us to move.<br />

The knee is a synovial weight-bearing joint, which comprises many tissues that maintain the<br />

mobile function; articular cartilage, <strong>subchondral</strong> <strong>bone</strong>, ligaments, capsule, synovial membrane and<br />

supporting muscles (fig. 1A). The ends <strong>of</strong> the adjoining <strong>bone</strong>s are covered with articular cartilage,<br />

which functions as a shock-absorber and provides a friction-free surface that enables the <strong>bone</strong>s<br />

to move smoothly against each other. The joint capsule, tendons, muscles and ligaments provide<br />

stability <strong>of</strong> the joint. The <strong>bone</strong>s are connected by strong ligaments and the tendons provide the<br />

attachment <strong>of</strong> muscles to the <strong>bone</strong>. The inside <strong>of</strong> the joint is covered with a synovial membrane,<br />

which provides and holds the lubricating synovial fluid that enables the joint to move easily (see<br />

fig. 1B) 13,14 .<br />

Fig. 1. Illustration <strong>of</strong> a healthy synovial knee joint. The different components <strong>of</strong> a knee<br />

joint are outlined in figure A and B. A) The components responsible for stability and<br />

movement are shown. B) The components <strong>of</strong> the articular capsule, which provide an easy<br />

friction, are shown in details. The figure was produced by Madsen, S.H.<br />

2.2.1 Development <strong>of</strong> <strong>bone</strong> from a cartilage template<br />

Although OA is a disease <strong>of</strong> the whole joint, this thesis focuses on <strong>bone</strong> and cartilage, which are<br />

the most affected tissues in OA. A description <strong>of</strong> the development <strong>of</strong> the joint, with respect to<br />

<strong>bone</strong> and cartilage, will help the reader to understand the tight connections <strong>between</strong> these two<br />

tissues.<br />

15


CHAPTER 2: Introduction<br />

Bone is a specialized tissue, which together with cartilage make up the human skeleton. The<br />

<strong>bone</strong>s have three main functions; 1) providing mechanical support for the whole body and acting<br />

as muscle attachment for locomotion, 2) providing protective shields for the vital organs and the<br />

<strong>bone</strong> marrow, and 3) providing a reserve for ions, mainly calcium and phosphate, and for growth<br />

factors and cytokines 15,16 .<br />

There are several subtypes <strong>of</strong> <strong>bone</strong>s; long <strong>bone</strong>, flat <strong>bone</strong>s, short <strong>bone</strong>s and<br />

irregular <strong>bone</strong>s 17 , <strong>of</strong> which only long <strong>bone</strong>s - e.g. femur and tibia, which constitute the knee - will<br />

be discussed further in this thesis. Long <strong>bone</strong>s are developed by endochondral ossification as<br />

shown in fig. 2A 18 . This whole process starts with a condensation <strong>of</strong> mesenchymal cells to form<br />

the cartilage anlage <strong>of</strong> the future <strong>bone</strong>. The mesenchymal cells further proliferate and<br />

differentiate into chondroblasts. The chondroblasts produce a cartilaginous matrix, where the<br />

predominant collagen is collagen type II. The chondroblasts become progressively embedded<br />

within their own matrix, where they lie within lacunae, and they are then called chondrocytes (the<br />

only cell type in articular cartilage). Eventually, the chondrocytes may further differentiate into<br />

hypertrophic chondrocytes whereby the cartilage becomes calcified before the hypertrophic<br />

chondrocytes undergo apoptosis and then disappear from the lacunae. This generates the primary<br />

ossification-centre, which splits into two opposite growth plates that moves from the centre <strong>of</strong><br />

the long <strong>bone</strong>, towards the ends. At this stage <strong>of</strong> <strong>bone</strong> development, the blood vessels penetrate<br />

the calcified matrix supplying osteoclast precursors. Differentiated osteoclasts (<strong>bone</strong> resorbing<br />

cells) use the lacunas in the calcified cartilage matrix as templates to initiate resorption.<br />

Fig. 2. Development <strong>of</strong> long <strong>bone</strong> by endochondral ossification. Schematic diagram showing the initial<br />

stages <strong>of</strong> endochondral ossification. A) Endochondral ossification is initiated from accumulated mesenchymal<br />

cells, which continue to differentiate and starts forming the primary, and then later the two secondary<br />

ossification centres. Trabecular <strong>bone</strong> is present after several cycles <strong>of</strong> remodelling 16 . B) The growth plate<br />

<strong>between</strong> the epiphysis and diaphysis is enlarged and divided in the different zones. The growth plate<br />

demonstrates the different stages <strong>of</strong> chondrocyte differentiation involved in endochondral <strong>bone</strong> formation.<br />

The figure was produced by Madsen, S.H.<br />

16


CHAPTER 2: Introduction<br />

Also osteoblasts (<strong>bone</strong> forming cells) arrive with the bloodstream and form a layer <strong>of</strong> woven<br />

<strong>bone</strong> on top <strong>of</strong> the remaining cartilage. The septae <strong>of</strong> calcified cartilage separating the lacunae are<br />

first resorbed by osteoclasts, while the remaining septae that extend into the diaphysis are used<br />

for osteoblasts to deposit <strong>bone</strong> matrix, resulting in the vascular <strong>bone</strong> marrow cavity (fig. 2A) 19 .<br />

The <strong>bone</strong> continues to grow through both the growth plates in appertaining epiphyses.<br />

In the growth plate (see fig. 2B) resting chondrocytes resume proliferation<br />

organized in columns toward the diaphysis and then mature into hypertrophic cells that express<br />

specific genes, such as collagen type X, and calcify the matrix. After apoptosis <strong>of</strong> the<br />

hypertrophic chondrocytes, the calcified matrix is resorbed by osteoclasts followed by <strong>bone</strong><br />

deposit by osteoblasts. This process is known as the <strong>bone</strong> modelling process, where <strong>bone</strong><br />

resorption and formation occur independently <strong>of</strong> each other. This modelling continues as long as<br />

needed, and eventually, secondary ossification-centres begin to form at the epiphyseal ends <strong>of</strong> the<br />

long <strong>bone</strong>.<br />

At the end <strong>of</strong> human adolescents, the proliferation <strong>of</strong> chondrocytes in the growth<br />

plate slows down and eventually stops. The continuous replacement <strong>of</strong> cartilage by <strong>bone</strong> results<br />

in the obliteration <strong>of</strong> the growth plate. Only articular cartilage remains. During primary <strong>bone</strong><br />

formation, woven <strong>bone</strong> structures are first formed, which comprise some amount <strong>of</strong> calcified<br />

cartilage. The woven <strong>bone</strong> will later be replaced by the more robust lamellar <strong>bone</strong> without<br />

calcified cartilage, during remodelling <strong>of</strong> the <strong>bone</strong> matrix (described in section 2.2.3), which is a<br />

different process than the modelling process. Mineralization <strong>of</strong> articular cartilage and its<br />

replacement by <strong>bone</strong> continues in the adult, however, at a much reduced rate than in growing<br />

<strong>bone</strong> 19,20 .<br />

2.2.2 Adult <strong>bone</strong> - macroscopic and microscopic organization<br />

After a long <strong>bone</strong> is fully developed, it has two epiphyses covered with a layer <strong>of</strong> articular<br />

cartilage, a cylindrical hollow portion in the middle called the diaphysis, and a transition zone<br />

<strong>between</strong> them called the metaphysis (see fig. 3) 16 .<br />

The long <strong>bone</strong> is divided in two types <strong>of</strong> structural <strong>bone</strong> (fig. 3):<br />

I) The external part <strong>of</strong> the long <strong>bone</strong> is formed by a thick and dense layer <strong>of</strong> calcified tissue called<br />

the cortex, which encloses the medullary cavity where the hematopoietic <strong>bone</strong> marrow is<br />

housed 17 .<br />

II) Toward the metaphysis and the epiphysis, the cortex becomes progressively thinner and the<br />

internal space is filled with a network <strong>of</strong> thin, calcified trabecular <strong>bone</strong>. The spaces enclosed by<br />

these thin trabeculae are also filled with hematopoietic <strong>bone</strong> marrow and are continuous with the<br />

diaphyseal medullary cavity 17 .<br />

17


CHAPTER 2: Introduction<br />

18<br />

Fig. 3. Schematic drawing <strong>of</strong> a long<br />

<strong>bone</strong>. A femur <strong>bone</strong> divided in sections:<br />

epiphyses, metaphyses, and a diaphysis.<br />

The different types <strong>of</strong> <strong>bone</strong> are shown;<br />

cortex (hard) and trabecular (spongy)<br />

<strong>bone</strong>. The proximal epiphysis (femur<br />

head) and the distal epiphysis are<br />

covered with cartilage to provide a<br />

smooth friction (during movement). The<br />

figure was produced by Madsen, S.H.<br />

The weight bearing <strong>bone</strong>s, such as the femur and tibia that comprises the knee, have two<br />

opposite and <strong>important</strong> functions. The first is to resist loading, thus the <strong>bone</strong> must be stiff. The<br />

second is to be flexible to withstand shock, compression and tension by energy absorption and<br />

deformation 21 . The stiffness is achieved by mineralization and flexibility is achieved by a suitable<br />

organic matrix composition. However, if the <strong>bone</strong> matrix is too mineralized, it will become brittle<br />

and more prone to fractures, whereas if it is under-mineralized, the <strong>bone</strong> will be too flexible and<br />

will crack more readily. Therefore, the functionality <strong>of</strong> the <strong>bone</strong> highly depends on the ratio <strong>of</strong><br />

organic and inorganic matrices 21 .<br />

In mammalian <strong>bone</strong>, the inorganic part <strong>of</strong> the matrix (50-70%) consist <strong>of</strong><br />

hydroxyapatite crystals [3Ca 3(PO 4) 2·(OH) 2], which are layered on and within collagen fibres,<br />

ensuring <strong>bone</strong> matrix stiffness (mineralization). The crystals tend to orient in the same direction<br />

as the collagen fibres. The organic part <strong>of</strong> the matrix (20-40%) consists mainly <strong>of</strong> collagen type I<br />

(approximately 90%), but also glycoproteins, proteoglycans, and non-collagenous proteins. The<br />

collagen type I molecule is a triple helix (described further in section 2.3.4), which is assembled<br />

into fibres, providing the tensile strength <strong>of</strong> the matrix. The orientation <strong>of</strong> the collagen fibres<br />

determines the microscopic structure <strong>of</strong> the <strong>bone</strong>. During <strong>bone</strong> development or fracture healing,<br />

where <strong>bone</strong> is formed very rapidly, there is no preferential organization <strong>of</strong> the collagen fibres; this<br />

type <strong>of</strong> <strong>bone</strong> is called woven <strong>bone</strong> (fig. 4) 17 . Woven <strong>bone</strong> is characterized by irregular bundles <strong>of</strong><br />

collagen fibres and calcification, which occur in irregularly distributed patches. Woven <strong>bone</strong> is<br />

progressively replaced by mature lamellar <strong>bone</strong> (fig. 4) during the remodelling process that


CHAPTER 2: Introduction<br />

follows normal development or healing. Lamellar <strong>bone</strong> is characterized by organized fibres,<br />

which are forming arches for optimal <strong>bone</strong> strength. The lamellae can be parallel to each other if<br />

deposited along a flat surface (trabecular <strong>bone</strong>) or concentric if deposited on a surface<br />

surrounding a channel centred on a blood vessel 17,22,23 .<br />

2.2.3 The cells <strong>of</strong> <strong>bone</strong> and their communications<br />

19<br />

Fig. 4. Lamellar and woven <strong>bone</strong>.<br />

Histological cut from fibula <strong>of</strong> rabbit<br />

showing details <strong>of</strong> lamellar <strong>bone</strong><br />

concentrically organized and woven<br />

<strong>bone</strong> mixed with cartilage and calcified<br />

cartilage tissues 24 .<br />

Bone contains several types <strong>of</strong> cells, which are <strong>important</strong> during development, repair, and<br />

maintenance. During <strong>bone</strong> development, the woven <strong>bone</strong> is modelled by resorption <strong>of</strong> calcified<br />

cartilage by osteoclasts and formation <strong>of</strong> <strong>bone</strong> by osteoblasts, independently <strong>of</strong> each other (not<br />

directly coupled) 16,25,26 . This is different in adult <strong>bone</strong>, where the tissue is constantly being<br />

remodelled by a communication/coupling <strong>between</strong> osteoclasts and osteoblasts, which enable<br />

<strong>bone</strong> to regenerate old or damaged tissue itself, adapt to changing stress environments, and<br />

control calcium homeostasis.<br />

Activation <strong>of</strong> the <strong>bone</strong> remodelling cycle is believed to be initiated by the<br />

osteocytes 15,21,27,28 , which comprises the most abundant cell type in <strong>bone</strong>. Osteocytes are cells<br />

trapped in the <strong>bone</strong> matrix. They rest in fluid-filled lacunae and communicate with surrounding<br />

cells via extensions called canaliculi 23,29 . When <strong>bone</strong> ages, the tissue eventually suffers<br />

microdamage and the surrounding osteocytes undergo apoptosis, which leads to signalling to the<br />

nearest <strong>bone</strong> lining cells (fig. 5A). This results in increased expression <strong>of</strong> receptor activator <strong>of</strong><br />

nuclear factor kappa-B ligand (RANKL), which attracts osteoclast precursors to the damaged<br />

area and initiates osteoclastogenesis. Osteoclasts derived from hematopoietic stem cells that are<br />

differentiated into monocytes and subsequently pre-osteoclasts, which then fuse into large<br />

multinucleated osteoclasts. The two cytokines, macrophage colony stimulating factor (M-CSF)<br />

and RANKL, are produced by the cells <strong>of</strong> the osteoblast lineage, and are very <strong>important</strong> for the<br />

differentiation <strong>of</strong> the osteoclasts (fig. 5A).


CHAPTER 2: Introduction<br />

20<br />

Fig. 5. The remodelling cycle on trabecular<br />

<strong>bone</strong>. A) A microcrack or old <strong>bone</strong> causes<br />

osteocytic apoptosis, which induces RANKL<br />

expression by <strong>bone</strong> lining cells. This attracts<br />

monocytes to the damaged <strong>bone</strong> area, and<br />

osteoclastogenesis starts. The osteoclasts will<br />

start to resorb <strong>bone</strong>, which is followed by<br />

formation <strong>of</strong> new <strong>bone</strong> matrix by the osteoblasts.<br />

Osteoblasts that are trapped in the remodelled<br />

matrix become osteocytes, whereas the rest<br />

either die or become flattened osteoblast lining<br />

cells. B) The resorbing osteoclast seals tightly to<br />

the <strong>bone</strong> surface to create a closed local<br />

environment known as the resorption lacuna.<br />

This is where the <strong>bone</strong> resorption takes place, by<br />

lowering the pH (secretion <strong>of</strong> H + ) and by<br />

releasing proteases by exocytosis. The figures<br />

were produced by Madsen, S.H.<br />

Active osteoclasts can be characterized by their ruffled cell borders, which seal and degrade the<br />

underlying <strong>bone</strong> in the resorption pit, through the release <strong>of</strong> hydrogen ions (lowering <strong>of</strong> pH) and<br />

proteases (such as cathepsin K and matrix metalloproteases). The resorbed <strong>bone</strong> tissue is<br />

transported across the osteoclast in small vesicles and is released into the extracellular fluid (fig.<br />

5B.) 21 .


CHAPTER 2: Introduction<br />

When the job is done, the osteoclasts undergo apoptosis, and osteoblast precursors are attracted<br />

to the site and are differentiated into mature osteoblasts. Osteoblasts are mononucleated <strong>bone</strong><br />

forming cells, which derive from mesenchymal stem cells that are differentiated into<br />

osteoprogenitor cell (in the presence <strong>of</strong> <strong>bone</strong> morphogenic protein (BMP)), and further into pre-<br />

osteoblasts (fig. 5A). The mature osteoblasts deposit osteoid in which the collagens (mainly<br />

collagen type I) are layered in an orderly fashion at sites <strong>of</strong> resorption, which converts into<br />

lamellar <strong>bone</strong> when mineralized (a function also executed by the osteoblasts) 30,31,32,33,34 . When an<br />

adequate amount <strong>of</strong> <strong>bone</strong> has been formed, the <strong>bone</strong> remodelling cycle ceases, and the<br />

osteoblasts trapped in the <strong>bone</strong> matrix become osteocytes. The remaining cells undergo<br />

apoptosis or differentiate into <strong>bone</strong> lining cells (fig. 5A) 27,35,36 .<br />

The remodelling process showed in fig. 5A outlines <strong>important</strong> communication<br />

processes from the osteoclasts to the osteoblasts, which is known as the coupling process that<br />

ensures that resorbed <strong>bone</strong> is replaced by an equal amount <strong>of</strong> new <strong>bone</strong> 35,36,37,38 . There are several<br />

theories about how this coupling process is regulated. These includes; the action <strong>of</strong> factors<br />

released from the <strong>bone</strong> during resorption 27,35,36,39 , a closed <strong>bone</strong> remodelling<br />

compartment 25,40,41,42,43 , a secretion <strong>of</strong> factors from the osteoclasts themselves or surface<br />

signalling 35,44,45,46 . Reversely, osteoblasts are very <strong>important</strong> for the osteoclastogenesis, as they are<br />

the source <strong>of</strong> M-CSF, RANKL and osteoprotegerin (OPG), in which the latter is a soluble decoy<br />

receptor for RANKL 31,33,47 . Further, in the normal regulation <strong>of</strong> calcium homeostasis, parathyroid<br />

hormone (PTH) stimulates osteoblasts to release RANKL and decreases the level <strong>of</strong> OPG. The<br />

increased level <strong>of</strong> RANKL then stimulates osteoclasts to resorb <strong>bone</strong> 26,48 . These findings support<br />

the conclusion that diverse and complex signalling systems coordinate the communication<br />

<strong>between</strong> the <strong>bone</strong> cells.<br />

The communication <strong>between</strong> osteoclasts and osteoclasts<br />

is evident. The signal to osteoclasts through PTH via<br />

osteoblasts is clear, whereas the coupling from<br />

osteoclasts to osteoblasts is still debated.<br />

No matter what... there is a communication <strong>between</strong><br />

the different cell types!<br />

21


2.2.4 Articular cartilage<br />

CHAPTER 2: Introduction<br />

In a normal knee joint, the epiphyses <strong>of</strong> femur and tibia are covered with a layer <strong>of</strong> articular<br />

cartilage, which functions as shock-absorber and provides a friction-free surface that enables the<br />

<strong>bone</strong>s to move smoothly against each other. Articular cartilage is avascular and is not innervated.<br />

In this way, nutrients from the synovial fluid are transported to the chondrocytes (the only cell<br />

type in cartilage), and waste products are transported back to the synovial fluid by diffusion.<br />

Chondrocytes are cartilage specific and comprise 2-5% <strong>of</strong> the total cartilage 5,11,14,49 . Their function<br />

is to produce and maintain the surrounding matrix. Hence, the physical condition <strong>of</strong> cartilage<br />

ultimately depends on the health <strong>of</strong> these cells. Similar to osteoblasts, chondrocytes derive from<br />

the mesenchymal stem cells 19,50 , and they differentiate into pre-hypertrophic and hypertrophic<br />

chondrocytes before they undergo apoptosis. Even though chondrocytes are metabolically very<br />

active, they normally do not divide after adolescence. Only small defects associated with minimal<br />

loss <strong>of</strong> matrix components are regenerated. More extensive defects exceed the repair capacity,<br />

and consequently the damage becomes permanent 51,52,53 .<br />

Cartilage tissue has unique viscoelastic and compressive properties provided by the<br />

extracellular matrix (ECM) (fig. 6), which consists mainly <strong>of</strong> collagen type II (15%) and forms a<br />

fibrous network that entraps proteoglycans (10%), <strong>of</strong> which aggrecan is the predominant<br />

molecule. The aggrecan molecule is a protein that covalently binds sulphated glycosaminoglycan<br />

(sGAG) chains to the core protein, which is non-covalently attached to hyaluronic acid. The<br />

aggrecans attract water molecules, due to the high negatively charged sGAGs, and allow the<br />

tissue to withstand compressive forces (water comprises approximately 70% <strong>of</strong><br />

cartilage) 5,14,49,54,55,56 . The ECM also contains a number <strong>of</strong> less abundant molecules such as minor<br />

collagens, growth factors and adhesive molecules, which are out <strong>of</strong> the scope <strong>of</strong> this thesis 14 .<br />

22<br />

Fig. 6. Cartilage components.<br />

The articular cartilage is mainly<br />

composed <strong>of</strong> collagen type II,<br />

chondrocytes and aggrecan<br />

attached to hyaluronic acid. The<br />

figure was produced by<br />

Madsen, S.H.


CHAPTER 2: Introduction<br />

Cartilage is divided into different zones that have different chondrocyte phenotypes,<br />

compositions, structures, and biomechanical properties (fig. 7). From the articular cartilage<br />

surface and down to the <strong>subchondral</strong> <strong>bone</strong>, we find the superficial zone, the middle zone, the<br />

deep zone and the calcified cartilage zone (fig. 7A). Since the superficial zone is exposed to<br />

compression, tensile forces and diffusion, it has flattened chondrocytes and is rich in collagen<br />

fibres that are parallel arranged to the surface (fig. 7B). In the middle zone, chondrocytes have a<br />

spherical shape and are randomly distributed. The collagen fibres are arranged in an angulated<br />

manner at which the compression load is better dispersed. The deep zone is responsible for<br />

providing the greatest resistance to compressive forces. The collagen fibres are arranged in a<br />

radial disposition, with the highest proteoglycan content, and the lowest water concentration. The<br />

chondrocytes are still spherical, but are typically arranged in columnar orientation parallel to the<br />

collagen fibres. The tide mark distinguishes the deep zone from the calcified cartilage zone. The<br />

calcified layer plays an integral role in securing the cartilage to <strong>bone</strong> by anchoring the collagen<br />

fibrils <strong>of</strong> the deep zone to the <strong>subchondral</strong> <strong>bone</strong>. In the calcified zone, the cell population is<br />

scarce and chondrocytes are hypertrophic 57,58 .<br />

Fig. 7. Schematic illustration <strong>of</strong> the zones observed within<br />

articular cartilage. A) 3D view <strong>of</strong> articular cartilage divided in<br />

different zones. B) The arrangements <strong>of</strong> collagens in the different<br />

zones are illustrated in the boxes next to the cartilage zones. Adapted<br />

with modifications from Sah et al. 59 .<br />

23


CHAPTER 2: Introduction<br />

In healthy mature cartilage, the normal turnover <strong>of</strong> the ECM is kept in equilibrium by cytokines<br />

and growth factors, which stimulate the production <strong>of</strong> proteinases and matrix components.<br />

Chondrocytes maintain a low turnover rate with collagen having a half-life <strong>of</strong> more than 100<br />

years and aggrecan having a half-life ranging from 3-24 years. In OA, the turnover is moved<br />

toward more degradation than formation <strong>of</strong> the cartilage 11,14 .<br />

Chondrocytes and osteoblasts both derive from<br />

the mesenchymal stem cells, suggesting a<br />

possible connection or natural communication<br />

<strong>between</strong> the cells.<br />

24


2.3 The pathology <strong>of</strong> Osteoarthritis<br />

CHAPTER 2: Introduction<br />

OA is a complex disease <strong>of</strong> the entire joint. It is mainly characterized by the progressive<br />

degradation <strong>of</strong> articular cartilage, formation <strong>of</strong> osteophytes (<strong>bone</strong> outgrowths) and alterations and<br />

thickening <strong>of</strong> the <strong>subchondral</strong> <strong>bone</strong> (fig. 8) 60,61 . The main focus in the past decades has been on<br />

the articular cartilage as the affected tissue and biomechanics as the causative agent. The<br />

alterations in <strong>subchondral</strong> <strong>bone</strong> were believed to be secondary to the enforced inactivity.<br />

However, several studies have shown that normal cartilage turnover is dependent on a healthy<br />

<strong>subchondral</strong> <strong>bone</strong> 62,63 . Today, it is openly debated whether the pathogenesis <strong>of</strong> OA originates in<br />

the <strong>bone</strong> or in the cartilage compartment, and it is not clear to which extend these two<br />

compartments drives disease progression. An increased amount <strong>of</strong> evidence suggests a strong<br />

coupling <strong>between</strong> the <strong>subchondral</strong> <strong>bone</strong> and the articular cartilage turnover with pathological<br />

processes occurring concurrently in both compartments. Therefore, an optimal intervention<br />

strategy for OA likely includes the targeting <strong>of</strong> both <strong>bone</strong> and cartilage compartments that affects<br />

the pathophysiology <strong>of</strong> the whole joint by modulating and altering the <strong>interactions</strong> <strong>between</strong> the<br />

different cell types.<br />

Fig. 8. Illustration <strong>of</strong> a knee with severe osteoarthritis.<br />

A severe chronic osteoarthritic knee, with eroded cartilage,<br />

eroded meniscus, exposed <strong>bone</strong> and osteophytes (<strong>bone</strong><br />

outgrowths). Adapted with few modifications 64 .<br />

25


CHAPTER 2: Introduction<br />

A schematic overview <strong>of</strong> the histopathological progression <strong>of</strong> OA is shown in fig. 9 65 .<br />

In very early OA, the superficial layer <strong>of</strong> cartilage is lost, the <strong>subchondral</strong> <strong>bone</strong> is getting<br />

denser, and blood vessels are penetrating the calcified cartilage.<br />

This affects the rest <strong>of</strong> the cartilage zones, which lose their organized matrix structures<br />

(fibrillation and loss <strong>of</strong> proteoglycans) and chondrocyte order (cluster formation).<br />

Simultaneously, the tidemark duplicates and is penetrated by blood vessels, and the<br />

<strong>subchondral</strong> <strong>bone</strong> increases in size and density as the cartilage is calcified and replaced by<br />

new <strong>bone</strong>.<br />

In moderate OA, the organization <strong>of</strong> the collagen network and chondrocyte column<br />

organization are lost and some <strong>of</strong> the chondrocytes have differentiated into hypertrophic<br />

chondrocytes. The <strong>subchondral</strong> <strong>bone</strong> still increases in size and penetrate the cartilage<br />

compartment.<br />

In late OA, only some parts <strong>of</strong> the deep zone <strong>of</strong> cartilage is left and the <strong>subchondral</strong> <strong>bone</strong> has<br />

increased in size and intermingled with cartilage (calcified zone has disappeared).<br />

At the end stage OA, the cartilage is lost and the <strong>subchondral</strong> <strong>bone</strong> is exposed. This may lead<br />

to synthesis <strong>of</strong> woven <strong>bone</strong>, which results in deformation <strong>of</strong> the joint.<br />

Fig. 9. The pathological development <strong>of</strong> OA. The progression <strong>of</strong> OA is shown from the healthy stage to the<br />

end stage <strong>of</strong> the disease, with focus on the matrix structure, cell number and phenotype, and ratio <strong>between</strong><br />

cartilage and <strong>subchondral</strong> <strong>bone</strong>. This figure illustrates the simultaneous processes <strong>of</strong> both <strong>bone</strong> and cartilage in<br />

the pathogenesis <strong>of</strong> OA. Figure adapted from Bay-Jensen et al. 65 .<br />

2.3.1 A chondrocyte is not just a chondrocyte<br />

Most people develop OA in later stages <strong>of</strong> life. One <strong>of</strong> the main reasons lies within the adult<br />

articular chondrocytes, which are ultimately responsible for remodelling and maintaining the<br />

health <strong>of</strong> the cartilage, even though these cells possess only little regenerative capacity. Research<br />

has shown that chondrocyte proliferation is rare in normal adult cartilage, and a reduction in the<br />

26


CHAPTER 2: Introduction<br />

number <strong>of</strong> active chondrocytes occurs with age along with a reduction in their response to<br />

anabolic factors. Therefore, the increased loss <strong>of</strong> chondrocytes during the later stages <strong>of</strong> life and<br />

thus the increasing lack <strong>of</strong> regeneration eventually result in damaged cartilage and the<br />

development <strong>of</strong> OA 66,67,68,69,70 . However, in the pathogenesis <strong>of</strong> OA, chondrocytes proliferate to<br />

form multiple cells within a chondron, which is a pivotal change from normal cartilage, where<br />

more than 2–3 cells are unusual, even in the elderly 71 . These clusters <strong>of</strong> chondrocytes in OA are<br />

not to be mistaken for the chondrocytes in healthy adult cartilage, as chondrocytes can change<br />

into different phenotypes during the progression <strong>of</strong> OA:<br />

An anabolic phenotype that expresses ECM proteins; e.g. collagen type II and aggrecan 72<br />

A catabolic phenotype that expresses proteolytic enzymes that degrade the ECM 11,73,74<br />

A Hypertrophic phenotype that expresses collagen type X before it undergoes apoptosis 75<br />

A dedifferentiated phenotype that expresses collagen type I (<strong>of</strong>ten in repair areas after damage 76,77<br />

A chondroblastic phenotype that expresses fetal type IIA collagen and type III collagen 78<br />

The differentiation <strong>of</strong> chondrocytes into these phenotypes probably occurs due to a “panic-<br />

attack” from the insufficient adult chondrocytes, who cannot keep up with the repair<br />

mechanisms <strong>of</strong> damaged cartilage. As a solution, the very productive phenotypes seen from the<br />

skeletal development are starting to emerge. Evidence <strong>of</strong> phenotypic change for chondrocytes is<br />

reflected in the presence <strong>of</strong> collagens not normally found in adult articular cartilage. As an<br />

example, the splice-variant <strong>of</strong> collagen type II, type IIA (which is normally expressed by<br />

chondroblasts during development) is re-expressed in early and late-stage OA 78 . Other indicators<br />

<strong>of</strong> a potential reversion <strong>of</strong> the cells to an earlier developmental phenotype are the hypertrophic<br />

chondrocyte marker, collagen type X 79 as well as the general increased production <strong>of</strong> proteins,<br />

proteinases, growth factors, cytokines, and other inflammatory mediators by more<br />

active/productive chondrocytes 80,81,82 .<br />

The adult articular chondrocytes normally maintain the cartilage<br />

homeostasis with a low turnover <strong>of</strong> matrix constituents. In OA,<br />

the chondrocytes attempt to recapitulate phenotypes <strong>of</strong> early stages<br />

<strong>of</strong> cartilage development, whose rate <strong>of</strong> synthesis <strong>of</strong> matrix<br />

constituents are faster. However, and unfortunately, the original<br />

cartilage cannot be replicated.<br />

27


2.3.2 The extracellular matrix <strong>of</strong> cartilage in OA<br />

CHAPTER 2: Introduction<br />

The health <strong>of</strong> normal cartilage is maintained by a balance <strong>between</strong> matrix synthesis and<br />

degradation. This balance is disturbed in OA. In early OA, there is increased synthesis <strong>of</strong> the<br />

principal matrix molecules <strong>of</strong> cartilage 82 , including collagen type II and aggrecan. However, the<br />

increased levels <strong>of</strong> these matrix molecules may be exported to the synovial fluid rather than<br />

incorporated into the tissue 56,83 . This is seen by an elevated rate <strong>of</strong> cartilage proteoglycan turnover<br />

in emerging OA and observed in the serum as an increase <strong>of</strong> aggrecan fragments in the synovial<br />

fluid and sulphate epitopes 84 . Thus, there is a decrease in total proteoglycan concentration, chain<br />

length and aggregation in OA cartilage 85 .<br />

Proteoglycans play an <strong>important</strong> role in keeping the water in<br />

the matrix. Thus it is <strong>important</strong> to understand the<br />

degradation processes <strong>of</strong> aggrecan in OA, to find the right<br />

treatment for the right stages <strong>of</strong> OA. This thesis investigates<br />

the different degradations processes for aggrecan (PAPER III)<br />

The elevated matrix synthesis in OA is accompanied by increased synthesis <strong>of</strong> proteases and<br />

other inflammatory factors, which may be related to elevated levels <strong>of</strong> interleukin-1 (IL-1) and<br />

tumour necrosis factor α (TNF-α) 86,87 . Multiple proteases are involved in the degradation <strong>of</strong><br />

cartilage, including cysteine-, aspartic-, serine-, and metalloproteinases. Especially, the expression<br />

<strong>of</strong> the zinc-dependent matrix metalloproteinase (MMP) family <strong>of</strong> enzymes is highly up-regulated<br />

in OA cartilage 88,89 . Mainly, MMP-1, -2, -3, -8, -9, -13 and -14 play a major part in protein<br />

degradation. MMP-1, -8 and -13 (collagenases) mainly cleave collagen type II in the triple<br />

helix 81,90,91,92 , whereas MMP-2 and -9 (gelatinases) cleave in the N-teleopeptide end 93 . Most MMPs<br />

are secreted by chondrocytes as latent proMMPs, which can be activated by other proteases<br />

through cleavage. As an example, MMP-2, -3 and membrane bound MMP-14 cleave other<br />

proenzymes (e.g. MMP-13) 94,95 . MMP-3 (stromelysin) also breaks down collagen type II and<br />

proteoglycans 96,97 . The breakdown <strong>of</strong> proteoglycans is also mediated by the aggrecanases, a<br />

disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS), and this breakdown<br />

is thought to be an early step in the loss <strong>of</strong> cartilage in OA 98 .<br />

The activity <strong>of</strong> MMPs and ADAMTS are normally controlled by the endogenous<br />

tissue inhibitors <strong>of</strong> metalloproteinases (TIMPs) 99 , which are also expressed by chondrocytes. An<br />

imbalance <strong>between</strong> metalloproteinases and TIMPs, results in cartilage degradation 89,100 . The<br />

expression <strong>of</strong> these TIMPs is down regulated by TNF-α in OA 101 , which furthermore contributes<br />

to the imbalance <strong>between</strong> metalloproteinases and their inhibitors.<br />

28


CHAPTER 2: Introduction<br />

Cysteine proteases are also essential players in the proteolytic cleavage <strong>of</strong> ECM in OA, which are<br />

synthesized both by the chondrocytes and synovial cells in response to cytokines and growth<br />

factors 102 . Cysteine proteases are normally responsible for the breakdown <strong>of</strong> collagen in <strong>bone</strong>,<br />

especially collagen type I 89 . However, cysteine proteases, like cathepsins K, B and L, have also<br />

been implicated as major participants in the degradation <strong>of</strong> collagen type II and<br />

proteoglycans 103,104,105,106 . As an example, expression <strong>of</strong> cathepsin K is up regulated in articular<br />

chondrocytes in a transgenic mouse model for OA 102 and urine concentrations <strong>of</strong> the cartilage<br />

breakdown products <strong>of</strong> collagen type II (CTX-II) are reduced by chronic cathepsin K<br />

inhibition 107 . Furthermore, expression <strong>of</strong> cathepsin B, H, L, and S are seen in hypertrophic<br />

chondrocytes, which are a hallmark <strong>of</strong> OA 108 . Besides the above mentioned actions, the main role<br />

<strong>of</strong> cathepsins is believed to be indirect degradation <strong>of</strong> matrix proteins through activation <strong>of</strong><br />

MMPs 109,110,111 .<br />

2.3.3 The <strong>subchondral</strong> <strong>bone</strong> in OA<br />

The <strong>subchondral</strong> <strong>bone</strong> turnover increases in OA 5,112 , which is demonstrated by the increase in<br />

alkaline phosphatase, osteocalcin, MMP expression (MMP-2 and -3), cytokines (TNF-α, IL-1, IL-<br />

8, IL-10), transforming growth factor beta (TGF-β) and TIMP-1 113,114 . Furthermore, the<br />

thickening <strong>of</strong> the <strong>subchondral</strong> <strong>bone</strong> with an abnormally low mineralization pattern 115 supports a<br />

greater proportion <strong>of</strong> osteoid in the diseased tissue with a higher turnover rate 112 . These changes<br />

in mineralization and structural organization, <strong>of</strong> the <strong>subchondral</strong> <strong>bone</strong> compartment, result in<br />

tissue stiffness. This stiffness impairs the ability <strong>of</strong> the <strong>subchondral</strong> <strong>bone</strong> to act as shock absorber<br />

to the overlying cartilage 116 .<br />

As described in section 2.1, the etiology <strong>of</strong> OA is unknown. An increasing line <strong>of</strong><br />

evidence suggests that both <strong>bone</strong> and cartilage, and not cartilage alone, contribute to the onset<br />

and progression <strong>of</strong> the disease. Several studies have addressed the importance <strong>of</strong> <strong>subchondral</strong><br />

<strong>bone</strong> in the pathogenesis <strong>of</strong> OA. In a study with a primate model <strong>of</strong> spontaneous OA, the<br />

progression <strong>of</strong> the disease severity <strong>of</strong> cartilage correlated with the increased thickness <strong>of</strong> the<br />

<strong>subchondral</strong> <strong>bone</strong>. Moreover, the <strong>bone</strong> changes preceded the changes in cartilage 117 . Another<br />

study showed a correlation <strong>between</strong> cartilage loss in OA patients and increased <strong>subchondral</strong> <strong>bone</strong><br />

turnover as measured by uptake <strong>of</strong> technetium-labelled bisphosphonate 118 . These studies confirm<br />

the importance <strong>of</strong> <strong>subchondral</strong> <strong>bone</strong>, either as an initiator or as a “partner in crime” in the<br />

pathogenesis <strong>of</strong> OA.<br />

The knowledge <strong>of</strong> <strong>subchondral</strong> <strong>bone</strong> in OA is limited. Thus this area<br />

needs to be investigated more thoroughly, as both tissues are involved in<br />

the pathogenesis <strong>of</strong> OA. This thesis also focus on the communication<br />

<strong>between</strong> the cartilage and <strong>subchondral</strong> <strong>bone</strong> (PAPER I and II)<br />

29


CHAPTER 2: Introduction<br />

2.3.4 Diagnostics - biochemical markers as a tool<br />

Several methods may be used to detect OA. X-ray detection is the golden standard method, where<br />

narrowing <strong>of</strong> the joint space indicates cartilage loss. Other ways to detect cartilage damage include<br />

arthroscopy and magnetic resonance imaging (MRI) 54,119 . However, OA is <strong>of</strong>ten diagnosed at late<br />

stages when the pain is unbearable for the patients, and the damages are too severe to repair.<br />

Today, many biochemical markers have been developed for the purpose <strong>of</strong> diagnosing the stage or<br />

degree <strong>of</strong> a specific disease.<br />

A biochemical marker is a single molecule or fragment <strong>of</strong> a molecule that is released<br />

into biological fluids (e.g. blood and urine) during tissue pathogenesis 54 . An increase in a specific<br />

biochemical marker indicates alterations <strong>of</strong> turnover, i.e., increased synthesis or breakdown 120 .<br />

Biochemical markers give a more accurate diagnosis <strong>of</strong> the severity <strong>of</strong> a disease for each patient, so<br />

that treatment could be designed specifically for the individual patient. Furthermore, the response<br />

to treatment or slow-down <strong>of</strong> the disease-progression can also be detected with these biochemical<br />

markers.<br />

OA is a disease affecting the whole joint and potential biochemical markers could<br />

originate from cartilage, <strong>bone</strong>, synovium, ligaments, tendons and muscles. However, the systemic<br />

levels from muscles, and some <strong>of</strong> the other tissues may exceed the small alterations seen from the<br />

OA-joint. At present, clinical studies indicate that different biochemical markers <strong>of</strong> cartilaginous<br />

matrix proteins may be useful to predict the early stages <strong>of</strong> cartilage damage and progression in<br />

OA. However, detection <strong>of</strong> OA by biochemical markers alone is not possible yet, as they measure<br />

systemic levels 54 . Nevertheless, the market for developing biochemical markers is increasing as it is<br />

possible to design more target-specific biochemical markers (e.g. neo-epitopes with post-<br />

translational modifications) or combine a specific selection <strong>of</strong> markers that represent one unique<br />

disease 121,122 .<br />

In OA, a central hallmark is the gradual destruction <strong>of</strong> articular cartilage and<br />

alterations in the <strong>subchondral</strong> <strong>bone</strong> 121 . The turnover <strong>of</strong> <strong>subchondral</strong> <strong>bone</strong> increases in early OA,<br />

which can be measured by increased formation and degradation <strong>of</strong> collagen type I (fig. 10A) and<br />

osteocalcin 123,124 . The resorption <strong>of</strong> collagen type I by osteoclasts is mediated by the protease,<br />

cathepsin K, which generates the neo-epitope, C-terminal telopeptide <strong>of</strong> collagen type I<br />

(CTX-I) 125,126,127 . However, studies have shown that both CTX-I and osteocalcin decrease in the<br />

body fluids <strong>of</strong> OA patients, indicating that overall <strong>bone</strong> turnover decreases in OA, whereas the<br />

increased turnover seen in <strong>subchondral</strong> <strong>bone</strong> is lost in systemic levels 128,129 .<br />

The turnover <strong>of</strong> cartilage is normally maintained by a balance <strong>between</strong> catabolic and<br />

anabolic processes; however, in the case <strong>of</strong> pathological matrix destruction, the rate <strong>of</strong> cartilage<br />

degradation exceeds the rate <strong>of</strong> formation, resulting in a net loss <strong>of</strong> cartilage matrix. During<br />

degradation <strong>of</strong> articular cartilage, MMPs and aggrecanases are considered the most <strong>important</strong><br />

proteases for degradation <strong>of</strong> articular cartilage 130,131,132 . The key target proteins in cartilage is<br />

collagen type II and aggrecan (fig. 10B+C). Other <strong>important</strong> molecules in articular cartilage<br />

30


CHAPTER 2: Introduction<br />

include cartilage oligomeric matrix protein (COMP), link protein and hyaluronic acid, which are<br />

out <strong>of</strong> the scope <strong>of</strong> the thesis.<br />

Fig. 10. Schematic illustration <strong>of</strong> proteolytic cleavage sites on collagen type I, collagen type II, and<br />

aggrecan. A) Collagen type I from <strong>bone</strong>. Formation <strong>of</strong> collagen type I is measured by PINP and PICP.<br />

Degradation <strong>of</strong> collagen type I by osteoclasts through cathepsin K cleavage is measured by CTX-I. B) Collagen<br />

type II from cartilage. Formation <strong>of</strong> collagen type II is measured by PIINP, PIIANP, and PIICP. Degradation <strong>of</strong><br />

collagen type II by MMP cleavage is measured by CTX-II or CIIM. C) Aggrecan from cartilage. Aggrecan<br />

degradation is measured by the MMP-mediated marker, 342-G2, and the aggrecanase-mediated marker, 374-G2.<br />

The sGAGs (red lines) are also measureable as a marker, representing aggrecan turnover. The three figures were<br />

produced by Madsen, S.H.<br />

31


CHAPTER 2: Introduction<br />

The degradation and formation <strong>of</strong> collagen type II can be measured by several biochemical<br />

markers 133 , but the present thesis mainly focuses on the MMP-mediated biochemical markers; e.g.<br />

C-terminal telopeptide <strong>of</strong> collagen type II (CTX-II) 129,134 and the novel CIIM 135 (fig. 10B). Several<br />

studies have shown that the CTX-II level is elevated in OA patients and in cartilage degradation<br />

from ex vivo-cultures 134,136,137 . It remains to be resolved whether this increase in CTX-II results from<br />

degradation <strong>of</strong> pre-existing collagen type II or newly synthesized collagen 138,139 . The formation<br />

markers <strong>of</strong> collagen type II, PIINP or PIIANP can in combination with the CTX-II or CIIM<br />

markers provide a more complete understanding <strong>of</strong> the turnover-actions. PIIANP is an attractive<br />

formation marker for OA-cartilage as it is an indicator <strong>of</strong> expression <strong>of</strong> the fetal form <strong>of</strong> collagen<br />

type IIA, which is re-expressed by OA chondrocytes 78,140 . The formation markers, PIINP and<br />

PIIANP, are normally suppressed in OA patients 141,142 . Another study has shown that PIIANP<br />

levels are increased in OA patients 138 , suggesting that PIIANP values are not always consistent.<br />

The degradation <strong>of</strong> aggrecan has attracted attention as a biochemical marker, as<br />

aggrecan degradation occurs early in OA and prior to collagen degradation 143 . Both MMP and<br />

aggrecanase-cleaved aggrecan fragments have been identified in synovial fluid 144 . The<br />

concentration <strong>of</strong> aggrecanase-mediated fragments (374-G2) dominated the MMP-mediated<br />

fragments (342-G2) in cartilage degradation ex vivo 144,145 , suggesting that the major mediators <strong>of</strong><br />

aggrecan loss in OA are the aggrecanases.<br />

Biochemical markers are key-tools used to<br />

investigate the hypotheses in this thesis.<br />

32


2.4 The effect <strong>of</strong> Glucocorticoids<br />

CHAPTER 2: Introduction<br />

Glucocorticoids (GCs) belongs to a class <strong>of</strong> steroid hormones that bind to the glucocorticoid<br />

receptor (GR). This receptor is present in almost every vertebrate animal cell and activation <strong>of</strong><br />

GRs initiate an up-regulation <strong>of</strong> the expression <strong>of</strong> anti-inflammatory proteins and represses the<br />

expression <strong>of</strong> pro-inflammatory proteins 146 . Consequently, GCs have potent anti-inflammatory<br />

and immunosuppressive properties and are thus part <strong>of</strong> the feedback mechanism in the immune<br />

system that decreases the inflammation 147,148 . Therefore, GCs are used as drugs to treat<br />

inflammatory conditions such as arthritis (e.g. rheumatoid arthritis), dermatitis, and diseases that<br />

are caused by an overactive immune system (allergies, asthma, and autoimmune diseases) (Table<br />

1) 146,148 . GCs are also administered as post-transplant immunosuppressants to prevent any acute<br />

transplant rejection and graft-versus-host disease. However, they do not prevent an infection and<br />

they also inhibit later reparative processes 146 .<br />

Treatment with GCs has many diverse effects, including potentially harmful side<br />

effects (Table 1) 146 . Excessive GC levels resulting from administration as a drug affect many<br />

systems, including inhibition <strong>of</strong> <strong>bone</strong> formation, suppression <strong>of</strong> calcium absorption and delayed<br />

wound healing 148 . With respect to OA, intra-articular injections <strong>of</strong> GCs appear to have a reducing<br />

effect on inflammation and pain 149 . Unfortunately, prolonged GC therapy results in GC-induced<br />

osteoporosis 150 . Thus, to treat OA, GCs without detrimental effects on <strong>bone</strong> are needed.<br />

The major endogenous GC in humans is cortisol, which was first used<br />

therapeutically for rheumatoid arthritis by Hench and co-workers in 1949 151 . Since then, a large<br />

number <strong>of</strong> synthetic compounds with GC activity have been developed for therapeutic use. They<br />

differ in pharmacokinetics and pharmacodynamics 152 . The potencies and effects <strong>of</strong> some <strong>of</strong> the<br />

synthetic glucocorticoids used for treatment are described in Table 1.<br />

Table 1. Synthetic glucocorticoids and their effects/side-effects. Most <strong>of</strong> the synthetic glucocorticoids have<br />

the same general effect (anti-inflammatory and immunosuppressive effects), but the potency and specificity for<br />

certain diseases vary 153,152 .<br />

33


CHAPTER 2: Introduction<br />

Hopefully, future studies may better define the molecular mechanisms <strong>of</strong> GC actions, thus<br />

triggering the discovery <strong>of</strong> new GC formulations providing a better pharmacokinetic pr<strong>of</strong>ile<br />

and fewer adverse effects.<br />

As GCs may be a potential treatment for<br />

OA, the effects <strong>of</strong> prednisolone and<br />

dexamethasone on <strong>bone</strong> and cartilage cells<br />

are investigated in PAPER II<br />

34


2.5 References for CHAPTER 2<br />

1 WebMD. Better information. Better health.<br />

http://www.webmd.com/rheumatoidarthritis/guide/most-common-arthritis-types.<br />

2011;<br />

2 Gigtforeningen.<br />

www.gigtforeningen.dk/viden+om+gigt/diagnoser/slidgi<br />

gt. 2011;<br />

3 Wu C.W. and Kalunian K.C. New developments in<br />

osteoarthritis. Clin Geriatr Med. 2005;21(3):589-601, vii.<br />

4 Lawrence R.C., Helmick C.G., Arnett F.C., Deyo R.A.,<br />

Felson D.T., Giannini E.H., Heyse S.P. et al. Estimates <strong>of</strong><br />

the prevalence <strong>of</strong> arthritis and selected musculoskeletal<br />

disorders in the United States. Arthritis Rheum.<br />

1998;41(5):778-799.<br />

5 Moskowitz R.W., Altman R.D., Hochberg M.C.,<br />

Buckwalter J.A., and Goldberg V.M. Chapter 3.<br />

Osteoarthritis; Diagnosis and Medical/surgical<br />

Management. 2007;4th(3)<br />

6 Cooper C., Cook P.L., Osmond C., Fisher L., and Cawley<br />

M.I. Osteoarthritis <strong>of</strong> the hip and osteoporosis <strong>of</strong> the<br />

proximal femur. Ann Rheum Dis. 1991;50(8):540-542.<br />

7 Foss M.V. and Byers P.D. Bone density, osteoarthrosis <strong>of</strong><br />

the hip, and fracture <strong>of</strong> the upper end <strong>of</strong> the femur. Ann<br />

Rheum Dis. 1972;31(4):259-264.<br />

8 Hilal G., Martel-Pelletier J., Pelletier J.P., Ranger P., and<br />

Lajeunesse D. Osteoblast-like cells from human<br />

<strong>subchondral</strong> osteoarthritic <strong>bone</strong> demonstrate an altered<br />

phenotype in vitro: possible role in <strong>subchondral</strong> <strong>bone</strong><br />

sclerosis. Arthritis Rheum. 1998;41(5):891-899.<br />

9 Dequeker J., Aerssens J., and Luyten F.P. Osteoarthritis<br />

and osteoporosis: clinical and research evidence <strong>of</strong> inverse<br />

relationship. Aging Clin Exp Res. 2003;15(5):426-439.<br />

10 Brandt K.D., Doherty M., and Lomander L.S.<br />

Osteoarthritis. 2003;<br />

11 Goldring M.B. Update on the biology <strong>of</strong> the chondrocyte<br />

and new approaches to treating cartilage diseases. Best<br />

Pract Res Clin Rheumatol. 2006;20(5):1003-1025.<br />

12 Felson D.T., Lawrence R.C., Dieppe P.A., Hirsch R.,<br />

Helmick C.G., Jordan J.M., Kington R.S. et al.<br />

Osteoarthritis: new insights. Part 1: the disease and its risk<br />

factors. Ann Intern Med. 2000;133(8):635-646.<br />

13 Worrall J.G. Understanding Arthritis and Rheumatism.<br />

2005;<br />

CHAPTER 2: Introduction<br />

35<br />

14 Rowan A.D. Cartilage catabolism in arthritis: factors that<br />

influence homeostasis. Expert Rev Mol Med. 2001;2001:1-<br />

20.<br />

15 Burr D.B. Targeted and nontargeted remodeling. Bone.<br />

2002;30(1):2-4.<br />

16 Baron R, Olsen B.J, Lian J.B, Canalis E., Monroe D.G.,<br />

Robey P.G., Mundy G.R. et al. Anatomy and Biology <strong>of</strong><br />

Bone Matrix and Cellular Elements. Primer on the<br />

Metabolic Bone Diseases and Disorders <strong>of</strong> Mineral<br />

Metabolism. 2003;5:1-58.<br />

17 Clarke B. Normal <strong>bone</strong> anatomy and physiology. Clin J<br />

Am Soc Nephrol. 2008;3 Suppl 3:S131-S139.<br />

18 Erlebacher A., Filvar<strong>of</strong>f E.H., Gitelman S.E., and Derynck<br />

R. Toward a molecular understanding <strong>of</strong> skeletal<br />

development. Cell. 1995;80(3):371-378.<br />

19 Keraplis A.C. Principles <strong>of</strong> <strong>bone</strong> biology. 2002;33-58.<br />

20 Provot S. and Schipani E. Molecular mechanisms <strong>of</strong><br />

endochondral <strong>bone</strong> development. Biochem Biophys Res<br />

Commun. 2005;328(3):658-665.<br />

21 Seeman E. and Delmas P.D. Bone quality--the material<br />

and structural basis <strong>of</strong> <strong>bone</strong> strength and fragility. N Engl<br />

J Med. 2006;354(21):2250-2261.<br />

22 Robey P.G. and Bodkey A.L. Extracellular matrix and<br />

biomineralization <strong>of</strong> <strong>bone</strong>. Primer on the metabolic <strong>bone</strong><br />

diseases and disorders <strong>of</strong> mineral metabolism.<br />

2003;5(6):38-46.<br />

23 Baron R. General principles <strong>of</strong> Bone Biology. Primer on<br />

the metabolic <strong>bone</strong> diseases and disorders <strong>of</strong> mineral<br />

metabolism. 2003;(1):1-8.<br />

24 Matos M.A., Araujo F.P., and Paixao F.B.<br />

Histomorphometric evaluation <strong>of</strong> <strong>bone</strong> healing in rabbit<br />

fibular osteotomy model without fixation. J Orthop Surg<br />

Res. 2008;3:4-<br />

25 Matsuo K. Cross-talk among <strong>bone</strong> cells. Curr Opin<br />

Nephrol Hypertens. 2009;18(4):292-297.<br />

26 Matsuo K. and Irie N. Osteoclast-osteoblast<br />

communication. Arch Biochem Biophys. 2008;473(2):201-<br />

209.<br />

27 Henriksen K., Neutzsky-Wulff A.V., Bonewald L.F., and<br />

Karsdal M.A. Local communication on and within <strong>bone</strong><br />

controls <strong>bone</strong> remodeling. Bone. 2009;44(6):1026-1033.


28 Iqbal J., Sun L., and Zaidi M. Coupling <strong>bone</strong> degradation<br />

to formation. Nat Med. 2009;15(7):729-731.<br />

29 Knothe Tate M.L., Adamson J.R., Tami A.E., and Bauer<br />

T.W. The osteocyte. Int J Biochem Cell Biol. 2004;36(1):1-<br />

8.<br />

30 Aubin J.E. Advances in the osteoblast lineage. Biochem<br />

Cell Biol. 1998;76(6):899-910.<br />

31 Gori F., H<strong>of</strong>bauer L.C., Dunstan C.R., Spelsberg T.C.,<br />

Khosla S., and Riggs B.L. The expression <strong>of</strong><br />

osteoprotegerin and RANK ligand and the support <strong>of</strong><br />

osteoclast formation by stromal-osteoblast lineage cells is<br />

developmentally regulated. Endocrinology.<br />

2000;141(12):4768-4776.<br />

32 Lerner U.H. NEW MOLECULES IN THE TUMOR<br />

NECROSIS FACTOR LIGAND AND RECEPTOR<br />

SUPERFAMILIES WITH IMPORTANCE FOR<br />

PHYSIOLOGICAL AND PATHOLOGICAL BONE<br />

RESORPTION. Crit Rev Oral Biol Med. 2004;15(2):64-<br />

81.<br />

33 Thomas G.P., Baker S.U., Eisman J.A., and Gardiner E.M.<br />

Changing RANKL/OPG mRNA expression in<br />

differentiating murine primary osteoblasts. J Endocrinol.<br />

2001;170(2):451-460.<br />

34 Teitelbaum S.L. Bone resorption by osteoclasts. Science.<br />

2000;289(5484):1504-1508.<br />

35 Martin T., Gooi J.H., and Sims N.A. Molecular<br />

mechanisms in coupling <strong>of</strong> <strong>bone</strong> formation to resorption.<br />

Crit Rev Eukaryot Gene Expr. 2009;19(1):73-88.<br />

36 Sims N.A. and Gooi J.H. Bone remodeling: Multiple<br />

cellular <strong>interactions</strong> required for coupling <strong>of</strong> <strong>bone</strong><br />

formation and resorption. Semin Cell Dev Biol.<br />

2008;19(5):444-451.<br />

37 Karsdal M.A., Henriksen K., Sorensen M.G., Gram J.,<br />

Schaller S., Dziegiel M.H., Heegaard A.M. et al.<br />

Acidification <strong>of</strong> the osteoclastic resorption compartment<br />

provides insight into the coupling <strong>of</strong> <strong>bone</strong> formation to<br />

<strong>bone</strong> resorption. Am J Pathol. 2005;166(2):467-476.<br />

38 Martin T.J. and Sims N.A. Osteoclast-derived activity in<br />

the coupling <strong>of</strong> <strong>bone</strong> formation to resorption. Trends Mol<br />

Med. 2005;11(2):76-81.<br />

39 Mundy G.R., Chen D., and Oyajobi B.O. Bone<br />

remodeling. Primer on the metabolic <strong>bone</strong> diseases and<br />

disorders <strong>of</strong> mineral metabolism. 2003;5(7):46-58.<br />

CHAPTER 2: Introduction<br />

36<br />

40 Andersen T.L., Sondergaard T.E., Skorzynska K.E., gnaes-<br />

Hansen F., Plesner T.L., Hauge E.M., Plesner T. et al. A<br />

physical mechanism for coupling <strong>bone</strong> resorption and<br />

formation in adult human <strong>bone</strong>. Am J Pathol.<br />

2009;174(1):239-247.<br />

41 Eriksen E.F., Eghbali-Fatourechi G.Z., and Khosla S.<br />

Remodeling and vascular spaces in <strong>bone</strong>. J Bone Miner<br />

Res. 2007;22(1):1-6.<br />

42 Hauge E.M., Qvesel D., Eriksen E.F., Mosekilde L., and<br />

Melsen F. Cancellous <strong>bone</strong> remodeling occurs in<br />

specialized compartments lined by cells expressing<br />

osteoblastic markers. J Bone Miner Res. 2001;16(9):1575-<br />

1582.<br />

43 Kaunitz J.D. and Yamaguchi D.T. TNAP, TrAP, ecto-<br />

purinergic signaling, and <strong>bone</strong> remodeling. J Cell Biochem.<br />

2008;105(3):655-662.<br />

44 Karsdal M.A., Martin T.J., Bollerslev J., Christiansen C.,<br />

and Henriksen K. Are nonresorbing osteoclasts sources <strong>of</strong><br />

<strong>bone</strong> anabolic activity? J Bone Miner Res. 2007;22(4):487-<br />

494.<br />

45 Karsdal M.A., Neutzsky-Wulff A.V., Dziegiel M.H.,<br />

Christiansen C., and Henriksen K. Osteoclasts secrete<br />

non-<strong>bone</strong> derived signals that induce <strong>bone</strong> formation.<br />

Biochem Biophys Res Commun. 2008;366(2):483-488.<br />

46 Zhao C., Irie N., Takada Y., Shimoda K., Miyamoto T.,<br />

Nishiwaki T., Suda T. et al. Bidirectional ephrinB2-<br />

EphB4 signaling controls <strong>bone</strong> homeostasis. Cell Metab.<br />

2006;4(2):111-121.<br />

47 Aubin J.E. and Bonnelye E. Osteoprotegerin and its<br />

ligand: a new paradigm for regulation <strong>of</strong><br />

osteoclastogenesis and <strong>bone</strong> resorption. Osteoporos Int.<br />

2000;11(11):905-913.<br />

48 Kwan T.S., Lajeunesse D., Pelletier J.P., and Martel-<br />

Pelletier J. Targeting <strong>subchondral</strong> <strong>bone</strong> for treating<br />

osteoarthritis: what is the evidence? Best Pract Res Clin<br />

Rheumatol. 2010;24(1):51-70.<br />

49 Mankin H.J., Brandt K.D., and Shulman L.E. Workshop<br />

on Etiopathogenesis <strong>of</strong> Osteoarthritis. The Journal <strong>of</strong><br />

Rheumatology. 1986;(13):1130-1160.<br />

50 Lefebvre V. and de C.B. Toward understanding SOX9<br />

function in chondrocyte differentiation. Matrix Biol.<br />

1998;16(9):529-540.


51 Lorenz H. and Richter W. Osteoarthritis: cellular and<br />

molecular changes in degenerating cartilage. Prog<br />

Histochem Cytochem. 2006;40(3):135-163.<br />

52 Wollheim F.A. Serum markers <strong>of</strong> articular cartilage<br />

damage and repair. Rheum Dis Clin North Am.<br />

1999;25(2):417-32, viii.<br />

53 Wotton S.F. and Duance V.C. Type III collagen in normal<br />

human articular cartilage. Histochem J. 1994;26(5):412-<br />

416.<br />

54 Charni-Ben T.N. and Garnero P. Monitoring cartilage<br />

turnover. Curr Rheumatol Rep. 2007;9(1):16-24.<br />

55 Hardingham T.E. and Fosang A.J. Proteoglycans: many<br />

forms and many functions. FASEB J. 1992;6(3):861-870.<br />

56 Matyas J.R., Adams M.E., Huang D., and Sandell L.J.<br />

Discoordinate gene expression <strong>of</strong> aggrecan and type II<br />

collagen in experimental osteoarthritis. Arthritis Rheum.<br />

1995;38(3):420-425.<br />

57 Darling E.M. and Athanasiou K.A. Rapid phenotypic<br />

changes in passaged articular chondrocyte subpopulations.<br />

J Orthop Res. 2005;23(2):425-432.<br />

58 Fox A.J.S, Bedi A., and Rodeo S.A. The Basic Science <strong>of</strong><br />

Articular Cartilage: Structure, Composition, and Function.<br />

Sports Health: A Multidisciplinary Approach.<br />

2009;1(6):461-461.<br />

59 Sah R.L, Wong B.L., Li K.W., Bae W.C, and Price J.H.<br />

Three dimentional histomorphometric analysis <strong>of</strong> normal<br />

adult human articular cartilage. 48th AMORS. 2001;<br />

60 Cao Y., Vacanti J.P., Paige K.T., Upton J., and Vacanti<br />

C.A. Transplantation <strong>of</strong> chondrocytes utilizing a polymercell<br />

construct to produce tissue-engineered cartilage in the<br />

shape <strong>of</strong> a human ear. Plast Reconstr Surg.<br />

1997;100(2):297-302.<br />

61 Poole A.R. An introduction to the pathophysiology <strong>of</strong><br />

osteoarthritis. Front Biosci. 1999;4:D662-D670.<br />

62 Hayami T., Pickarski M., Wesolowski G.A., McLane J.,<br />

Bone A., Destefano J., Rodan G.A. et al. The role <strong>of</strong><br />

<strong>subchondral</strong> <strong>bone</strong> remodeling in osteoarthritis: reduction<br />

<strong>of</strong> cartilage degeneration and prevention <strong>of</strong> osteophyte<br />

formation by alendronate in the rat anterior cruciate<br />

ligament transection model. Arthritis Rheum.<br />

2004;50(4):1193-1206.<br />

63 Casden A.M., Jaffe F.F., Kastenbaum D.M., and Bonar<br />

S.F. Osteoarthritis associated with osteopetrosis treated by<br />

total knee arthroplasty. Report <strong>of</strong> a case. Clin Orthop<br />

Relat Res. 1989;(247):202-207.<br />

CHAPTER 2: Introduction<br />

37<br />

64 www.zimmer.co.uk. 2011;<br />

65 Bay-Jensen A.C., Hoegh-Madsen S., Dam E., Henriksen<br />

K., Sondergaard B.C., Pastoureau P., Qvist P. et al. Which<br />

elements are involved in reversible and irreversible<br />

cartilage degradation in osteoarthritis? Rheumatol Int.<br />

2010;30(4):435-442.<br />

66 Vignon E., Arlot M., Patricot L.M., and Vignon G. The<br />

cell density <strong>of</strong> human femoral head cartilage. Clin Orthop<br />

Relat Res. 1976;(121):303-308.<br />

67 Horton W.E., Jr., Feng L., and Adams C. Chondrocyte<br />

apoptosis in development, aging and disease. Matrix Biol.<br />

1998;17(2):107-115.<br />

68 Hashimoto S., Takahashi K., Amiel D., Coutts R.D., and<br />

Lotz M. Chondrocyte apoptosis and nitric oxide<br />

production during experimentally induced osteoarthritis.<br />

Arthritis Rheum. 1998;41(7):1266-1274.<br />

69 Guerne P.A., Blanco F., Kaelin A., Desgeorges A., and<br />

Lotz M. Growth factor responsiveness <strong>of</strong> human articular<br />

chondrocytes in aging and development. Arthritis Rheum.<br />

1995;38(7):960-968.<br />

70 Martin J.A. and Buckwalter J.A. The role <strong>of</strong> chondrocyte<br />

senescence in the pathogenesis <strong>of</strong> osteoarthritis and in<br />

limiting cartilage repair. J Bone Joint Surg Am. 2003;85-A<br />

Suppl 2:106-110.<br />

71 Poole C.A., Ayad S., and Sch<strong>of</strong>ield J.R. Chondrons from<br />

articular cartilage: I. Immunolocalization <strong>of</strong> type VI<br />

collagen in the pericellular capsule <strong>of</strong> isolated canine tibial<br />

chondrons. J Cell Sci. 1988;90 ( Pt 4):635-643.<br />

72 Pfander D., Swoboda B., and Kirsch T. Expression <strong>of</strong><br />

early and late differentiation markers (proliferating cell<br />

nuclear antigen, syndecan-3, annexin VI, and alkaline<br />

phosphatase) by human osteoarthritic chondrocytes. Am J<br />

Pathol. 2001;159(5):1777-1783.<br />

73 Aigner T., Kurz B., Fukui N., and Sandell L. Roles <strong>of</strong><br />

chondrocytes in the pathogenesis <strong>of</strong> osteoarthritis. Curr<br />

Opin Rheumatol. 2002;14(5):578-584.<br />

74 Valcourt U., Gouttenoire J., ubert-Foucher E., Herbage<br />

D., and Mallein-Gerin F. Alternative splicing <strong>of</strong> type II<br />

procollagen pre-mRNA in chondrocytes is oppositely<br />

regulated by BMP-2 and TGF-beta1. FEBS Lett.<br />

2003;545(2-3):115-119.<br />

75 Kielty C.M., Kwan A.P., Holmes D.F., Schor S.L., and<br />

Grant M.E. Type X collagen, a product <strong>of</strong> hypertrophic<br />

chondrocytes. Biochem J. 1985;227(2):545-554.


76 Benya P.D. and Shaffer J.D. Dedifferentiated<br />

chondrocytes reexpress the differentiated collagen<br />

phenotype when cultured in agarose gels. Cell.<br />

1982;30(1):215-224.<br />

77 Elima K. and Vuorio E. Expression <strong>of</strong> mRNAs for<br />

collagens and other matrix components in<br />

dedifferentiating and redifferentiating human<br />

chondrocytes in culture. FEBS Lett. 1989;258(2):195-198.<br />

78 Aigner T., Zhu Y., Chansky H.H., Matsen F.A., III,<br />

Maloney W.J., and Sandell L.J. Reexpression <strong>of</strong> type IIA<br />

procollagen by adult articular chondrocytes in<br />

osteoarthritic cartilage. Arthritis Rheum. 1999;42(7):1443-<br />

1450.<br />

79 von der M.K., Kirsch T., Nerlich A., Kuss A., Weseloh G.,<br />

Gluckert K., and Stoss H. Type X collagen synthesis in<br />

human osteoarthritic cartilage. Indication <strong>of</strong> chondrocyte<br />

hypertrophy. Arthritis Rheum. 1992;35(7):806-811.<br />

80 Sandell L.J. and Aigner T. Articular cartilage and changes<br />

in arthritis. An introduction: cell biology <strong>of</strong> osteoarthritis.<br />

Arthritis Res. 2001;3(2):107-113.<br />

81 Tchetina E.V., Squires G., and Poole A.R. Increased type<br />

II collagen degradation and very early focal cartilage<br />

degeneration is associated with upregulation <strong>of</strong><br />

chondrocyte differentiation related genes in early human<br />

articular cartilage lesions. J Rheumatol. 2005;32(5):876-<br />

886.<br />

82 Goldring M.B. and Goldring S.R. Osteoarthritis. J Cell<br />

Physiol. 2007;213(3):626-634.<br />

83 Aigner T., Gluckert K., and von der M.K. Activation <strong>of</strong><br />

fibrillar collagen synthesis and phenotypic modulation <strong>of</strong><br />

chondrocytes in early human osteoarthritic cartilage<br />

lesions. Osteoarthritis Cartilage. 1997;5(3):183-189.<br />

84 Lohmander L.S., Ionescu M., Jugessur H., and Poole A.R.<br />

Changes in joint cartilage aggrecan after knee injury and in<br />

osteoarthritis. Arthritis Rheum. 1999;42(3):534-544.<br />

85 Bollet A.J. and Nance J.L. Biochemical Findings in<br />

Normal and Osteoarthritic Articular Cartilage. II.<br />

Chondroitin Sulfate Concentration and Chain Length,<br />

Water, and Ash Content. J Clin Invest. 1966;45(7):1170-<br />

1177.<br />

86 Goldring M.B. The role <strong>of</strong> the chondrocyte in<br />

osteoarthritis. Arthritis Rheum. 2000;43(9):1916-1926.<br />

87 Goldring M.B. Osteoarthritis and cartilage: the role <strong>of</strong><br />

cytokines. Curr Rheumatol Rep. 2000;2(6):459-465.<br />

CHAPTER 2: Introduction<br />

38<br />

88 Bramono D.S., Richmond J.C., Weitzel P.P., Kaplan D.L.,<br />

and Altman G.H. Matrix metalloproteinases and their<br />

clinical applications in orthopaedics. Clin Orthop Relat<br />

Res. 2004;(428):272-285.<br />

89 Cawston T.E. and Young D.A. Proteinases involved in<br />

matrix turnover during cartilage and <strong>bone</strong> breakdown. Cell<br />

Tissue Res. 2010;339(1):221-235.<br />

90 Hollander A.P., Pidoux I., Reiner A., Rorabeck C., Bourne<br />

R., and Poole A.R. Damage to type II collagen in aging<br />

and osteoarthritis starts at the articular surface, originates<br />

around chondrocytes, and extends into the cartilage with<br />

progressive degeneration. J Clin Invest. 1995;96(6):2859-<br />

2869.<br />

91 Shibakawa A., Yudoh K., Masuko-Hongo K., Kato T.,<br />

Nishioka K., and Nakamura H. The role <strong>of</strong> <strong>subchondral</strong><br />

<strong>bone</strong> resorption pits in osteoarthritis: MMP production by<br />

cells derived from <strong>bone</strong> marrow. Osteoarthritis Cartilage.<br />

2005;13(8):679-687.<br />

92 Bluteau G., Conrozier T., Mathieu P., Vignon E., Herbage<br />

D., and Mallein-Gerin F. Matrix metalloproteinase-1, -3, -<br />

13 and aggrecanase-1 and -2 are differentially expressed in<br />

experimental osteoarthritis. Biochim Biophys Acta.<br />

2001;1526(2):147-158.<br />

93 Freemont A.J., Hampson V., Tilman R., Goupille P.,<br />

Taiwo Y., and Hoyland J.A. Gene expression <strong>of</strong> matrix<br />

metalloproteinases 1, 3, and 9 by chondrocytes in<br />

osteoarthritic human knee articular cartilage is zone and<br />

grade specific. Ann Rheum Dis. 1997;56(9):542-549.<br />

94 Hanemaaijer R., Sorsa T., Konttinen Y.T., Ding Y.,<br />

Sutinen M., Visser H., van H., V et al. Matrix<br />

metalloproteinase-8 is expressed in rheumatoid synovial<br />

fibroblasts and endothelial cells. Regulation by tumor<br />

necrosis factor-alpha and doxycycline. J Biol Chem.<br />

1997;272(50):31504-31509.<br />

95 Salminen H.J., Saamanen A.M., Vankemmelbeke M.N.,<br />

Auho P.K., Perala M.P., and Vuorio E.I. Differential<br />

expression patterns <strong>of</strong> matrix metalloproteinases and their<br />

inhibitors during development <strong>of</strong> osteoarthritis in a<br />

transgenic mouse model. Ann Rheum Dis. 2002;61(7):591-<br />

597.<br />

96 Cole A.A. and Kuettner K.E. MMP-8 (neutrophil<br />

collagenase) mRNA and aggrecanase cleavage products<br />

are present in normal and osteoarthritic human articular<br />

cartilage. Acta Orthop Scand Suppl. 1995;266:98-102.


97 Nakamura H., Tanaka M., Masuko-Hongo K., Yudoh K.,<br />

Kato T., Beppu M., and Nishioka K. Enhanced<br />

production <strong>of</strong> MMP-1, MMP-3, MMP-13, and RANTES<br />

by interaction <strong>of</strong> chondrocytes with autologous T cells.<br />

Rheumatol Int. 2006;26(11):984-990.<br />

98 Neame P.J. and Sandy J.D. Cartilage aggrecan.<br />

Biosynthesis, degradation and osteoarthritis. J Fla Med<br />

Assoc. 1994;81(3):191-193.<br />

99 Smith R.L. Degradative enzymes in osteoarthritis. Front<br />

Biosci. 1999;4:D704-D712.<br />

100 Blanco F.J., Guitian R., Moreno J., de Toro F.J., and<br />

Galdo F. Effect <strong>of</strong> antiinflammatory drugs on COX-1 and<br />

COX-2 activity in human articular chondrocytes. J<br />

Rheumatol. 1999;26(6):1366-1373.<br />

101 Hui W., Rowan A.D., and Cawston T. Modulation <strong>of</strong> the<br />

expression <strong>of</strong> matrix metalloproteinase and tissue<br />

inhibitors <strong>of</strong> metalloproteinases by TGF-beta1 and IGF-1<br />

in primary human articular and bovine nasal chondrocytes<br />

stimulated with TNF-alpha. Cytokine. 2001;16(1):31-35.<br />

102 Morko J.P., Soderstrom M., Saamanen A.M., Salminen<br />

H.J., and Vuorio E.I. Up regulation <strong>of</strong> cathepsin K<br />

expression in articular chondrocytes in a transgenic mouse<br />

model for osteoarthritis. Ann Rheum Dis. 2004;63(6):649-<br />

655.<br />

103 Inaoka T., Bilbe G., Ishibashi O., Tezuka K., Kumegawa<br />

M., and Kokubo T. Molecular cloning <strong>of</strong> human cDNA<br />

for cathepsin K: novel cysteine proteinase predominantly<br />

expressed in <strong>bone</strong>. Biochem Biophys Res Commun.<br />

1995;206(1):89-96.<br />

104 Dodds R.A., Connor J.R., Drake F., Feild J., and Gowen<br />

M. Cathepsin K mRNA detection is restricted to<br />

osteoclasts during fetal mouse development. J Bone Miner<br />

Res. 1998;13(4):673-682.<br />

105 Kafienah W., Bromme D., Buttle D.J., Croucher L.J., and<br />

Hollander A.P. Human cathepsin K cleaves native type I<br />

and II collagens at the N-terminal end <strong>of</strong> the triple helix.<br />

Biochem J. 1998;331 ( Pt 3):727-732.<br />

106 Maciewicz R.A., Wotton S.F., Etherington D.J., and<br />

Duance V.C. Susceptibility <strong>of</strong> the cartilage collagens types<br />

II, IX and XI to degradation by the cysteine proteinases,<br />

cathepsins B and L. FEBS Lett. 1990;269(1):189-193.<br />

107 McDougall J.J., Schuelert N., and Bowyer J. Cathepsin K<br />

inhibition reduces CTXII levels and joint pain in the<br />

guinea pig model <strong>of</strong> spontaneous osteoarthritis.<br />

Osteoarthritis Cartilage. 2010;18(10):1355-1357.<br />

CHAPTER 2: Introduction<br />

39<br />

108 Uusitalo H., Hiltunen A., Soderstrom M., Aro H.T., and<br />

Vuorio E. Expression <strong>of</strong> cathepsins B, H, K, L, and S and<br />

matrix metalloproteinases 9 and 13 during chondrocyte<br />

hypertrophy and endochondral ossification in mouse<br />

fracture callus. Calcif Tissue Int. 2000;67(5):382-390.<br />

109 Knauper V., Will H., Lopez-Otin C., Smith B., Atkinson<br />

S.J., Stanton H., Hembry R.M. et al. Cellular mechanisms<br />

for human procollagenase-3 (MMP-13) activation.<br />

Evidence that MT1-MMP (MMP-14) and gelatinase a<br />

(MMP-2) are able to generate active enzyme. J Biol Chem.<br />

1996;271(29):17124-17131.<br />

110 Knauper V., Bailey L., Worley J.R., Soloway P., Patterson<br />

M.L., and Murphy G. Cellular activation <strong>of</strong> proMMP-13<br />

by MT1-MMP depends on the C-terminal domain <strong>of</strong><br />

MMP-13. FEBS Lett. 2002;532(1-2):127-130.<br />

111 Knauper V., Murphy G., and Tschesche H. Activation <strong>of</strong><br />

human neutrophil procollagenase by stromelysin 2. Eur J<br />

Biochem. 1996;235(1-2):187-191.<br />

112 Mansell J.P. and Bailey A.J. Abnormal cancellous <strong>bone</strong><br />

collagen metabolism in osteoarthritis. J Clin Invest.<br />

1998;101(8):1596-1603.<br />

113 Mansell J.P., Tarlton J.F., and Bailey A.J. Biochemical<br />

evidence for altered <strong>subchondral</strong> <strong>bone</strong> collagen<br />

metabolism in osteoarthritis <strong>of</strong> the hip. Br J Rheumatol.<br />

1997;36(1):16-19.<br />

114 Hulejova H., Baresova V., Klezl Z., Polanska M., Adam<br />

M., and Senolt L. Increased level <strong>of</strong> cytokines and matrix<br />

metalloproteinases in osteoarthritic <strong>subchondral</strong> <strong>bone</strong>.<br />

Cytokine. 2007;38(3):151-156.<br />

115 Grynpas M.D., Alpert B., Katz I., Lieberman I., and<br />

Pritzker K.P. Subchondral <strong>bone</strong> in osteoarthritis. Calcif<br />

Tissue Int. 1991;49(1):20-26.<br />

116 Radin E.L. and Rose R.M. Role <strong>of</strong> <strong>subchondral</strong> <strong>bone</strong> in<br />

the initiation and progression <strong>of</strong> cartilage damage. Clin<br />

Orthop Relat Res. 1986;(213):34-40.<br />

117 Carlson C.S., Loeser R.F., Purser C.B., Gardin J.F., and<br />

Jerome C.P. Osteoarthritis in cynomolgus macaques. III:<br />

Effects <strong>of</strong> age, gender, and <strong>subchondral</strong> <strong>bone</strong> thickness on<br />

the severity <strong>of</strong> disease. J Bone Miner Res. 1996;11(9):1209-<br />

1217.<br />

118 Dieppe P., Cushnaghan J., Young P., and Kirwan J.<br />

Prediction <strong>of</strong> the progression <strong>of</strong> joint space narrowing in<br />

osteoarthritis <strong>of</strong> the knee by <strong>bone</strong> scintigraphy. Ann<br />

Rheum Dis. 1993;52(8):557-563.


119 Menkes C.J. Radiographic criteria for classification <strong>of</strong><br />

osteoarthritis. J Rheumatol Suppl. 1991;27:13-15.<br />

120 Rousseau J.C. and Delmas P.D. Biological markers in<br />

osteoarthritis. Nat Clin Pract Rheumatol. 2007;3(6):346-<br />

356.<br />

121 Fosang A.J., Stanton H., Little C.B., and Atley L.M.<br />

Neoepitopes as biomarkers <strong>of</strong> cartilage catabolism.<br />

Inflamm Res. 2003;52(7):277-282.<br />

122 Karsdal M.A., Henriksen K., Leeming D.J., Woodworth<br />

T., Vassiliadis E., and Bay-Jensen A.C. Novel<br />

combinations <strong>of</strong> Post-Translational Modification (PTM)<br />

neo-epitopes provide tissue-specific biochemical markers--<br />

are they the cause or the consequence <strong>of</strong> the disease? Clin<br />

Biochem. 2010;43(10-11):793-804.<br />

123 Price P.A., Parthemore J.G., and Deftos L.J. New<br />

biochemical marker for <strong>bone</strong> metabolism. Measurement<br />

by radioimmunoassay <strong>of</strong> <strong>bone</strong> GLA protein in the plasma<br />

<strong>of</strong> normal subjects and patients with <strong>bone</strong> disease. J Clin<br />

Invest. 1980;66(5):878-883.<br />

124 Mouritzen U., Christgau S., Lehmann H.J., Tanko L.B.,<br />

and Christiansen C. Cartilage turnover assessed with a<br />

newly developed assay measuring collagen type II<br />

degradation products: influence <strong>of</strong> age, sex, menopause,<br />

hormone replacement therapy, and body mass index. Ann<br />

Rheum Dis. 2003;62(4):332-336.<br />

125 Bonde M., Qvist P., Fledelius C., Riis B.J., and<br />

Christiansen C. Immunoassay for quantifying type I<br />

collagen degradation products in urine evaluated. Clin<br />

Chem. 1994;40(11 Pt 1):2022-2025.<br />

126 Rosenquist C., Fledelius C., Christgau S., Pedersen B.J.,<br />

Bonde M., Qvist P., and Christiansen C. Serum CrossLaps<br />

One Step ELISA. First application <strong>of</strong> monoclonal<br />

antibodies for measurement in serum <strong>of</strong> <strong>bone</strong>-related<br />

degradation products from C-terminal telopeptides <strong>of</strong> type<br />

I collagen. Clin Chem. 1998;44(11):2281-2289.<br />

127 Schaller S., Henriksen K., Hoegh-Andersen P.,<br />

Sondergaard B.C., Sumer E.U., Tanko L.B., Qvist P. et al.<br />

In vitro, ex vivo, and in vivo methodological approaches<br />

for studying therapeutic targets <strong>of</strong> osteoporosis and<br />

degenerative joint diseases: how biomarkers can assist?<br />

Assay Drug Dev Technol. 2005;3(5):553-580.<br />

128 Garnero P., Rousseau J.C., and Delmas P.D. Molecular<br />

basis and clinical use <strong>of</strong> biochemical markers <strong>of</strong> <strong>bone</strong>,<br />

cartilage, and synovium in joint diseases. Arthritis Rheum.<br />

2000;43(5):953-968.<br />

CHAPTER 2: Introduction<br />

40<br />

129 Garnero P., Piperno M., Gineyts E., Christgau S., Delmas<br />

P.D., and Vignon E. Cross sectional evaluation <strong>of</strong><br />

biochemical markers <strong>of</strong> <strong>bone</strong>, cartilage, and synovial tissue<br />

metabolism in patients with knee osteoarthritis: relations<br />

with disease activity and joint damage. Ann Rheum Dis.<br />

2001;60(6):619-626.<br />

130 Bondeson J., Wainwright S., Hughes C., and Caterson B.<br />

The regulation <strong>of</strong> the ADAMTS4 and ADAMTS5<br />

aggrecanases in osteoarthritis: a review. Clin Exp<br />

Rheumatol. 2008;26(1):139-145.<br />

131 Zhen E.Y., Brittain I.J., Laska D.A., Mitchell P.G., Sumer<br />

E.U., Karsdal M.A., and Duffin K.L. Characterization <strong>of</strong><br />

metalloprotease cleavage products <strong>of</strong> human articular<br />

cartilage. Arthritis Rheum. 2008;58(8):2420-2431.<br />

132 Bay-Jensen A.C., Sondergaard B.C., Christiansen C.,<br />

Karsdal M.A., Madsen S.H., and Qvist P. Biochemical<br />

markers <strong>of</strong> joint tissue turnover. Assay Drug Dev<br />

Technol. 2010;8(1):118-124.<br />

133 Qvist P., Christiansen C., Karsdal M.A., Madsen S.H.,<br />

Sondergaard B.C., and Bay-Jensen A.C. Application <strong>of</strong><br />

biochemical markers in development <strong>of</strong> drugs for<br />

treatment <strong>of</strong> osteoarthritis. Biomarkers. 2010;15(1):1-19.<br />

134 Christgau S., Garnero P., Fledelius C., Moniz C., Ensig M.,<br />

Gineyts E., Rosenquist C. et al. Collagen type II C-<br />

telopeptide fragments as an index <strong>of</strong> cartilage degradation.<br />

Bone. 2001;29(3):209-215.<br />

135 Bay-Jensen A.C., Liu Q., Byrjalsen I., Li Y., Wang J.,<br />

Pedersen C., Leeming D.J. et al. Enzyme-linked<br />

immunosorbent assay (ELISAs) for metalloproteinase<br />

derived type II collagen neoepitope, CIIM--increased<br />

serum CIIM in subjects with severe radiographic<br />

osteoarthritis. Clin Biochem. 2011;44(5-6):423-429.<br />

136 Garnero P., Charni N., Juillet F., Conrozier T., and<br />

Vignon E. Increased urinary type II collagen helical and C<br />

telopeptide levels are independently associated with a<br />

rapidly destructive hip osteoarthritis. Ann Rheum Dis.<br />

2006;65(12):1639-1644.<br />

137 Sondergaard B.C., Henriksen K., Wulf H., Oestergaard S.,<br />

Schurigt U., Brauer R., Danielsen I. et al. Relative<br />

contribution <strong>of</strong> matrix metalloprotease and cysteine<br />

protease activities to cytokine-stimulated articular cartilage<br />

degradation. Osteoarthritis Cartilage. 2006;14(8):738-748.<br />

138 Sharif M., Kirwan J., Charni N., Sandell L.J., Whittles C.,<br />

and Garnero P. A 5-yr longitudinal study <strong>of</strong> type IIA<br />

collagen synthesis and total type II collagen degradation in<br />

patients with knee osteoarthritis--association with disease<br />

progression. Rheumatology (Oxford). 2007;46(6):938-943.


139 Garnero P., Ayral X., Rousseau J.C., Christgau S., Sandell<br />

L.J., Dougados M., and Delmas P.D. Uncoupling <strong>of</strong> type<br />

II collagen synthesis and degradation predicts progression<br />

<strong>of</strong> joint damage in patients with knee osteoarthritis.<br />

Arthritis Rheum. 2002;46(10):2613-2624.<br />

140 Aigner T., Stoss H., Weseloh G., Zeiler G., and von der<br />

M.K. Activation <strong>of</strong> collagen type II expression in<br />

osteoarthritic and rheumatoid cartilage. Virchows Arch B<br />

Cell Pathol Incl Mol Pathol. 1992;62(6):337-345.<br />

141 Nemirovskiy O.V., Sunyer T., Aggarwal P., Abrams M.,<br />

Hellio Le Graverand M.P., and Mathews W.R. Discovery<br />

and development <strong>of</strong> the N-terminal procollagen type II<br />

(NPII) biomarker: a tool for measuring collagen type II<br />

synthesis. Osteoarthritis Cartilage. 2008;16(12):1494-1500.<br />

142 Rousseau J.C., Zhu Y., Miossec P., Vignon E., Sandell L.J.,<br />

Garnero P., and Delmas P.D. Serum levels <strong>of</strong> type IIA<br />

procollagen amino terminal propeptide (PIIANP) are<br />

decreased in patients with knee osteoarthritis and<br />

rheumatoid arthritis. Osteoarthritis Cartilage.<br />

2004;12(6):440-447.<br />

143 Caterson B., Flannery C.R., Hughes C.E., and Little C.B.<br />

Mechanisms involved in cartilage proteoglycan catabolism.<br />

Matrix Biol. 2000;19(4):333-344.<br />

144 Sumer E.U., Qvist P., and Tanko L.B. Matrix<br />

Metalloproteinase and Aggrecanase Generated Aggrecan<br />

Fragments: Implications for the Diagnostics and<br />

Therapeutics <strong>of</strong> Destructive Joint Diseases. Drug<br />

development research. 2007;68(1):1-13.<br />

145 Sumer E.U., Sondergaard B.C., Rousseau J.C., Delmas<br />

P.D., Fosang A.J., Karsdal M.A., Christiansen C. et al.<br />

MMP and non-MMP-mediated release <strong>of</strong> aggrecan and its<br />

fragments from articular cartilage: a comparative study <strong>of</strong><br />

three different aggrecan and glycosaminoglycan assays.<br />

Osteoarthritis Cartilage. 2007;15(2):212-221.<br />

CHAPTER 2: Introduction<br />

41<br />

146 Glucocorticoid - wikipedia.<br />

http://en.wikipedia.org/wiki/Glucocorticoid. 2011;<br />

147 Rhen T. and Cidlowski J.A. Antiinflammatory action <strong>of</strong><br />

glucocorticoids--new mechanisms for old drugs. N Engl J<br />

Med. 2005;353(16):1711-1723.<br />

148 www.vivo.colostate.edu. 2011;<br />

149 Hepper C.T., Halvorson J.J., Duncan S.T., Gregory A.J.,<br />

Dunn W.R., and Spindler K.P. The efficacy and duration<br />

<strong>of</strong> intra-articular corticosteroid injection for knee<br />

osteoarthritis: a systematic review <strong>of</strong> level I studies. J Am<br />

Acad Orthop Surg. 2009;17(10):638-646.<br />

150 Eastell R., Reid D.M., Compston J., Cooper C., Fogelman<br />

I., Francis R.M., Hosking D.J. et al. A UK Consensus<br />

Group on management <strong>of</strong> glucocorticoid-induced<br />

osteoporosis: an update. J Intern Med. 1998;244(4):271-<br />

292.<br />

151 HENCH P.S., SLOCUMB C.H., BARNES A.R., SMITH<br />

H.C., POLLEY H.F., and KENDALL E.C. The effects <strong>of</strong><br />

the adrenal cortical hormone 17-hydroxy-11dehydrocorticosterone<br />

(Compound E) on the acute phase<br />

<strong>of</strong> rheumatic fever; preliminary report. Mayo Clin Proc.<br />

1949;24(11):277-297<br />

152 www.endotext.org/adrenal/adrenal14/adrenal14.htm.<br />

2011;<br />

153 Leung D.Y., Hanifin J.M., Charlesworth E.N., Li J.T.,<br />

Bernstein I.L., Berger W.E., Blessing-Moore J. et al.<br />

Disease management <strong>of</strong> atopic dermatitis: a practice<br />

parameter. Joint Task Force on Practice Parameters,<br />

representing the American Academy <strong>of</strong> Allergy, Asthma<br />

and Immunology, the American College <strong>of</strong> Allergy,<br />

Asthma and Immunology, and the Joint Council <strong>of</strong><br />

Allergy, Asthma and Immunology. Work Group on<br />

Atopic Dermatitis. Ann Allergy Asthma Immunol.<br />

1997;79(3):197-211.


Suzi Høgh Madsen, PhD thesis<br />

42


CHAPTER 3<br />

Overview <strong>of</strong> OA models<br />

CHAPTER 3: Overview <strong>of</strong> OA models<br />

This Overview aims to introduce some <strong>of</strong> the different pre-clinical models used today to<br />

investigate the pathogenesis <strong>of</strong> OA with emphasis on the ex vivo model. In brief, the complexity<br />

and advantages/disadvantages <strong>of</strong> the different models and the motivation for developing the<br />

femur head model (PAPER I) are described. Further, the controls used in previous ex vivo models<br />

are evaluated in order to understand the selection <strong>of</strong> these controls for the present femur head<br />

model (PAPER I).<br />

3.1 Pre-clinical models<br />

Clinical studies <strong>of</strong> OA in humans are expensive and very time consuming. Furthermore, clinical<br />

studies do not provide information about processes and mechanisms involved in pathogenic OA-<br />

tissues, which could be key information to prevent or stop the ongoing development <strong>of</strong> the<br />

disease. For these and ethical reasons, pre-clinical models are commonly used to investigate the<br />

metabolism <strong>of</strong> healthy/diseased <strong>bone</strong> or cartilage, which can be classified according to their<br />

complexity (fig. 11):<br />

In vitro; monolayer or pellet systems <strong>of</strong> isolated cells<br />

Ex vivo explants cultures, in which the cells are held in their natural matrix<br />

In vivo animal models; such as spontaneous, surgically induced, or transgenic, which resemble<br />

the human disease (apart from the species/origin)<br />

Each <strong>of</strong> these models has advantages and disadvantages depending on the purpose <strong>of</strong> the<br />

investigation. In the following section, the methodology behind the three models will be described<br />

in brief in relation to OA with main focus on the ex vivo model.<br />

Fig. 11. The different types <strong>of</strong> pre-clinical models. In an in vitro model, cells are isolated from their natural<br />

environments, which may cause them to change phenotype during culture. In an ex vivo model, the cells are<br />

maintained in their natural environments, but lack systemic influence (pressure, blood supply etc.). In in vivo<br />

models, the cells are maintained in their natural environment and host. The figure was produced by Madsen, S.H.<br />

43


CHAPTER 3: Overview <strong>of</strong> OA models<br />

3.2 Advantages/disadvantages <strong>of</strong> pre-clinical models<br />

3.2.1 In vitro<br />

Cultures <strong>of</strong> isolated chondrocytes are the easiest and most convenient way to investigate<br />

chondrogenesis in vitro; inexpensive, easy to setup and handle. However, several limitations must<br />

be considered. Two-dimensional monolayer cultures support the proliferation <strong>of</strong> articular<br />

chondrocytes, but change their phenotypic appearance to de-differentiated fibroblast-like cells,<br />

which synthesize collagen type I (a marker <strong>of</strong> osteoblasts) 1,2 . Consequently, cultures <strong>of</strong> isolated<br />

chondrocytes are not suitable for investigating cartilage degradation or for studying mechanisms<br />

involved in cartilage matrix assembly, but are rather used for investigating proteoglycan and<br />

collagen biosynthesis in response to various stimuli (Table 2) 3 . Pellet culture systems were<br />

originally described as a method for preventing the phenotypic de-differentiation <strong>of</strong> chondrocytes<br />

in vitro 4 . This culture system allows three-dimensional cell-cell interaction and is investigated by<br />

the same techniques as for isolated chondrocytes in monolayer, and additionally by<br />

histology/immunohistochemistry (Table 1) 5 .<br />

When investigating <strong>bone</strong> cells (osteoclasts and osteoblasts), the cells are not<br />

isolated from the <strong>bone</strong> matrix, but differentiated from their progenitor cells (e.g. isolated from<br />

blood) and cultured on e.g. <strong>bone</strong> or plastic 6 . Differently from isolated chondrocytes, osteoclasts<br />

and osteoblasts are able to resorb and deposit <strong>bone</strong>, respectively, which is their natural task in<br />

vivo. Thus, in vitro models for <strong>bone</strong> cells are very close to imitating in vivo conditions 6 . The<br />

osteoclast and osteoblast cell cultures have been used to evaluate the direct effect <strong>of</strong><br />

glucocorticoids in this thesis (PAPER II).<br />

3.2.2 Ex vivo<br />

The more complicated and technically advanced ex vivo explants model <strong>of</strong>fers the ability to<br />

investigate the biology <strong>of</strong> the tissue, including both ECM and cells. This model also <strong>of</strong>fers the<br />

ability to investigate how the tissue is affected by different compounds 7 . The cells are embedded<br />

in their natural matrix where the immediate environment is preserved, including macromolecules<br />

and cell-binding proteins. Additionally, the explants model allows preservation <strong>of</strong> the cell<br />

phenotype; e.g. the chondrocytes in cartilage explants maintain their spherical appearance (Table<br />

2) 8 . Several studies have used the cartilage explants model to resemble the pathological events in<br />

OA cartilage 9,10,11 . The pioneering work on porcine cartilage organ cultures were done by Fell et<br />

al. in 1973 12,13 . Since then, the cartilage explants model is widely used as a degradation assay that<br />

allows investigation <strong>of</strong> cartilage metabolism in response to different stimuli and agents 14,15,16 . A<br />

prominent example is that <strong>of</strong> Sondergaard et al., who showed that calcitonin abrogated the CTX-<br />

II release from catabolically induced cartilage explants 9 . Other studies investigated the proteolytic<br />

pathways in other cartilage ex vivo studies 10,17 . The advantage <strong>of</strong> studying cartilage metabolism in<br />

the cartilage explants model is the unique in vivo likeness. In combination with biochemical<br />

markers, this cartilage explants model is considered to be the best non-in vivo model in the study<br />

44


CHAPTER 3: Overview <strong>of</strong> OA models<br />

<strong>of</strong> OA 7,18 . The biochemical markers can be used in combination with histology/<br />

immunohistochemistry to further characterize the pathogenesis <strong>of</strong> the cartilage 14 . The articular<br />

cartilage explants model has been used in PAPER II, III, and IV to evaluate ECM turnover and<br />

enzymatic activity.<br />

There is an urgent need for the discovery and development <strong>of</strong> new treatments for<br />

OA. A fundamental step in discovering new therapies for OA is to critically evaluate our<br />

understanding <strong>of</strong> the etiology <strong>of</strong> the disease, including the complex relationship <strong>between</strong> cartilage<br />

and <strong>bone</strong>. Thus, the cartilage explants model is not complete as it only comprises cartilage. When<br />

the present PhD-project was initiated there were neither biological models nor tools that allowed<br />

the investigation <strong>of</strong> the interaction <strong>between</strong> the <strong>bone</strong> and cartilage cells. Thus, there was a strong<br />

need for an ex vivo model system comprising both <strong>bone</strong> and cartilage, which could provide a<br />

unique opportunity to investigate cell-cell <strong>interactions</strong> and test novel drug candidates for OA.<br />

This resulted in testing murine femur heads as potential candidates to constitute a novel explants<br />

model since the femur heads comprise both <strong>bone</strong> and cartilage (fig. 12). Importantly, the femur<br />

head is a closed system, which would not be damaged during isolation procedures (except on the<br />

shaft), keeping the homogeneity high <strong>between</strong> the samples, whereas the cartilage explants have a<br />

low homogeneity <strong>between</strong> the samples (Table 1). Isolation as a whole unit reduces the stress on<br />

the cells as the ECM is still intact and uncontrollable repair mechanisms are avoided.<br />

Fig. 12. The femur head comprises both <strong>bone</strong> and cartilage. The femur head is isolated from the femur<br />

by cutting with a scissor. The femur head is a whole unit, which is only damaged at the cutting site (at the<br />

femur neck). The femur head comprises both <strong>bone</strong> and cartilage, but also the transition zone <strong>of</strong><br />

<strong>bone</strong>/cartilage (e.g. growth plate during development). The <strong>interactions</strong> <strong>between</strong> the cells from the different<br />

compartments, primarily the chondrocytes, osteoclasts, and osteoblasts, can be investigated in this femur<br />

head explants model. The figure was produced by Madsen, S.H.<br />

The pathology <strong>of</strong> OA involves the whole joint. The<br />

development <strong>of</strong> a novel ex vivo model comprising both<br />

<strong>bone</strong> and cartilage is characterized in PAPER I<br />

45


3.2.3 In vivo<br />

CHAPTER 3: Overview <strong>of</strong> OA models<br />

Animal models are commonly used for investigating pathology that resembles the human disease<br />

<strong>of</strong> interest. These in vivo animal models can <strong>of</strong>ten follow preliminary in vitro or ex vivo studies (fig.<br />

11), which need their results to be confirmed in an in situ setting 7,19 . Furthermore, the complex<br />

changes in the joint-tissue <strong>of</strong> OA patients may take decades to develop and the need to<br />

investigate the early changes in robust and valid in vivo models has necessitated the use <strong>of</strong> living<br />

animals in experiments. A range <strong>of</strong> animal models <strong>of</strong> OA have been developed, such as<br />

spontaneous, surgically induced, and transgenic animal models. These models are not perfect, as<br />

species are not alike, but the models <strong>of</strong>fer the opportunity to display many <strong>of</strong> the pathologic<br />

features that characterize the human disease. For example, transgenic mice with mutations in<br />

collagen type II develops OA-like changes with age 20,21 , whereas surgically removed menisci can<br />

be observed to induce osteophyte formation, cartilage fibrillation, loss <strong>of</strong> proteoglycans, and<br />

cellular clustering within few weeks 22,23 . Moreover, several species spontaneously develop OA,<br />

which highly resembles human OA, although these models are relatively slow to develop the<br />

disease 24,25 .<br />

To summarize the advantages, animal models can be used to study pathologic<br />

events representing early OA in human patients, and they are applicable for investigation <strong>of</strong> the<br />

effects <strong>of</strong> a variety <strong>of</strong> agents on the progression <strong>of</strong> the disease. Furthermore, only animal models<br />

can show altered mental behaviour or catch potential side-effect from a drug (other tissues). One<br />

obvious disadvantage <strong>of</strong> the in vivo animal models is the potential differences in tissue response<br />

<strong>between</strong> animals and humans. Eventually, phase-trials <strong>of</strong> a potential drug would demonstrate the<br />

effect or lack <strong>of</strong> effect in humans. Another <strong>important</strong> disadvantage is the ethical aspect <strong>of</strong> using<br />

living animals, which will not be discussed further in this thesis. Below is a table with the<br />

different pre-clinical models <strong>of</strong> cartilage.<br />

Table 2. Comparison <strong>of</strong> different cartilage pre-clinical models. The advantages and disadvantages <strong>of</strong> the in<br />

vitro, ex vivo and in vivo models are outlined in the table, which makes it easy to compare to each other. Adapted<br />

with modifications from Sondergaard 26 .<br />

46


3.3 Ex vivo controls<br />

CHAPTER 3: Overview <strong>of</strong> OA models<br />

In order to design an ex vivo model for experimentation, it is critical to acquire appropriate<br />

controls, which induce an anabolic or catabolic system that represents regenerative or damaged<br />

tissue. The acknowledged cartilage explants model has established the best controls for cartilage:<br />

1) insulin-like growth factor-I (IGF-I) as an anabolic control, and 2) a combination <strong>of</strong> tumour<br />

necrosis factor-α (TNF-α) and oncostatin M (OSM) as a catabolic control 9,10,18 . In the early stages<br />

<strong>of</strong> OA, an increased amount <strong>of</strong> degrading enzymes is produced by the chondrocytes 27 . When<br />

articular cartilage explants are stimulated with TNF-α and OSM, the chondrocytes also produce<br />

these degrading enzymes 9 . OSM only induces a mild catabolic response in chondrocytes, but it<br />

act synergistically with TNF-α 10,28,29 . Thus, articular cartilage explants cultured in the presence <strong>of</strong><br />

OSM and TNF-α are shown to be a useful ex vivo model <strong>of</strong> OA 29 . Next is a short description <strong>of</strong><br />

the three key factors used in the cartilage explants model:<br />

IGF-I is <strong>important</strong> in skeletal development 30 , although this factor is primarily synthesized in the<br />

liver under the influence <strong>of</strong> growth hormones. IGF-I is essential for <strong>bone</strong> formation and it<br />

accounts for most <strong>of</strong> the chondrocyte-stimulating activity found in serum 30 . Several reports have<br />

described IGF-I to be the major anabolic factor <strong>of</strong> cartilage 31 . The ability <strong>of</strong> IGF-I to enhance<br />

matrix synthesis in normal cartilage is well established in vivo and in vitro 28,32 . For example, in<br />

cultures <strong>of</strong> human articular chondrocytes, IGF-I induces the production <strong>of</strong> GAGs and collagen<br />

type II 31 . Besides the chondroanabolic effects, it has been shown that IGF-I opposes the<br />

activities <strong>of</strong> catabolic cytokines in articular cartilage. Tyler showed in 1989 32 that IGF-I reduces<br />

proteoglycan degradation, probably by inhibiting the production and release <strong>of</strong> proteinases 28,33 .<br />

TNF-α is mainly produced by macrophages, but also by a broad variety <strong>of</strong> other cell types.<br />

TNF-α induces apoptosis, cellular proliferation, differentiation and inflammation. TNF-α has<br />

been shown to be an <strong>important</strong> mediator <strong>of</strong> cartilage destruction in vivo 28,32 . Besides mediating<br />

destruction <strong>of</strong> cartilage, TNF-α also inhibits synthesis <strong>of</strong> proteoglycan and collagen type II 27 .<br />

OSM is a member <strong>of</strong> the IL-6 family and is synthesized by stimulated T-cells and macrophages.<br />

Besides inhibition <strong>of</strong> proteoglycan synthesis, OSM also stimulates chondrocytes to produce<br />

proteinases, which induce proteoglycan degradation 27 . However, Wahl & Wallace indicated in<br />

2001 that OSM is anabolic; promoting wound healing, <strong>bone</strong> formation and anti-inflammatory<br />

effects 34 .<br />

The controls from the cartilage explants model<br />

were evaluated and used in the femur head<br />

explants model (PAPER I)<br />

47


3.4 References for CHAPTER 3<br />

1 von der M.K., Gauss V., von der M.H., and Muller P.<br />

Relationship <strong>between</strong> cell shape and type <strong>of</strong> collagen<br />

synthesised as chondrocytes lose their cartilage phenotype<br />

in culture. Nature. 1977;267(5611):531-532.<br />

2 Lee D.A., Reisler T., and Bader D.L. Expansion <strong>of</strong><br />

chondrocytes for tissue engineering in alginate beads<br />

enhances chondrocytic phenotype compared to<br />

conventional monolayer techniques. Acta Orthop Scand.<br />

2003;74(1):6-15.<br />

3 Takigawa M., Okawa T., Pan H., Aoki C., Takahashi K.,<br />

Zue J., Suzuki F. et al. Insulin-like growth factors I and II<br />

are autocrine factors in stimulating proteoglycan synthesis,<br />

a marker <strong>of</strong> differentiated chondrocytes, acting through<br />

their respective receptors on a clonal human<br />

chondrosarcoma-derived chondrocyte cell line, HCS-2/8.<br />

Endocrinology. 1997;138(10):4390-4400.<br />

4 Manning W.K. and Bonner W.M., Jr. Isolation and culture<br />

<strong>of</strong> chondrocytes from human adult articular cartilage.<br />

Arthritis Rheum. 1967;10(3):235-239.<br />

5 Mackay A.M., Beck S.C., Murphy J.M., Barry F.P.,<br />

Chichester C.O., and Pittenger M.F. Chondrogenic<br />

differentiation <strong>of</strong> cultured human mesenchymal stem cells<br />

from marrow. Tissue Eng. 1998;4(4):415-428.<br />

6 Neutzsky-Wulff A.V., Karsdal M.A., and Henriksen K.<br />

Characterization <strong>of</strong> the <strong>bone</strong> phenotype in ClC-7-deficient<br />

mice. Calcif Tissue Int. 2008;83(6):425-437.<br />

7 Schaller S., Henriksen K., Hoegh-Andersen P.,<br />

Sondergaard B.C., Sumer E.U., Tanko L.B., Qvist P. et al.<br />

In vitro, ex vivo, and in vivo methodological approaches<br />

for studying therapeutic targets <strong>of</strong> osteoporosis and<br />

degenerative joint diseases: how biomarkers can assist?<br />

Assay Drug Dev Technol. 2005;3(5):553-580.<br />

8 Hascall V.C., Morales T.I., Hascall G.K., Handley C.J.,<br />

and McQuillan D.J. Biosynthesis and turnover <strong>of</strong><br />

proteoglycans in organ culture <strong>of</strong> bovine articular cartilage.<br />

J Rheumatol Suppl. 1983;11:45-52.<br />

9 Sondergaard B.C., Wulf H., Henriksen K., Schaller S.,<br />

Oestergaard S., Qvist P., Tanko L.B. et al. Calcitonin<br />

directly attenuates collagen type II degradation by<br />

inhibition <strong>of</strong> matrix metalloproteinase expression and<br />

activity in articular chondrocytes. Osteoarthritis Cartilage.<br />

2006;14(8):759-768.<br />

CHAPTER 3: Overview <strong>of</strong> OA models<br />

48<br />

10 Sondergaard B.C., Henriksen K., Wulf H., Oestergaard S.,<br />

Schurigt U., Brauer R., Danielsen I. et al. Relative<br />

contribution <strong>of</strong> matrix metalloprotease and cysteine<br />

protease activities to cytokine-stimulated articular cartilage<br />

degradation. Osteoarthritis Cartilage. 2006;14(8):738-748.<br />

11 Charni-Ben T.N., Desmarais S., Bay-Jensen A.C., Delaisse<br />

J.M., Percival M.D., and Garnero P. The type II collagen<br />

fragments Helix-II and CTX-II reveal different enzymatic<br />

pathways <strong>of</strong> human cartilage collagen degradation.<br />

Osteoarthritis Cartilage. 2008;16(10):1183-1191.<br />

12 Dingle J.T., Horsfield P., Fell H.B., and Barratt M.E.<br />

Breakdown <strong>of</strong> proteoglycan and collagen induced in pig<br />

articular cartilage in organ culture. Ann Rheum Dis.<br />

1975;34(4):303-311.<br />

13 Fell H.B. and Barratt M.E. The role <strong>of</strong> s<strong>of</strong>t connective<br />

tissue in the breakdown <strong>of</strong> pig articular cartilage cultivated<br />

in the presence <strong>of</strong> complement-sufficient antiserum to pig<br />

erythrocytes. I. Histological changes. Int Arch Allergy<br />

Appl Immunol. 1973;44(3):441-468.<br />

14 Roy-Beaudry M., Martel-Pelletier J., Pelletier J.P., M'Barek<br />

K.N., Christgau S., Shipkolye F., and Moldovan F.<br />

Endothelin 1 promotes osteoarthritic cartilage degradation<br />

via matrix metalloprotease 1 and matrix metalloprotease<br />

13 induction. Arthritis Rheum. 2003;48(10):2855-2864.<br />

15 Saklatvala J. Tumour necrosis factor alpha stimulates<br />

resorption and inhibits synthesis <strong>of</strong> proteoglycan in<br />

cartilage. Nature. 1986;322(6079):547-549.<br />

16 Cawston T.E., Curry V.A., Summers C.A., Clark I.M.,<br />

Riley G.P., Life P.F., Spaull J.R. et al. The role <strong>of</strong><br />

oncostatin M in animal and human connective tissue<br />

collagen turnover and its localization within the<br />

rheumatoid joint. Arthritis Rheum. 1998;41(10):1760-<br />

1771.<br />

17 Sondergaard B.C., Schultz N., Madsen S.H., Bay-Jensen<br />

A.C., Kassem M., and Karsdal M.A. MAPKs are essential<br />

upstream signaling pathways in proteolytic cartilage<br />

degradation--divergence in pathways leading to<br />

aggrecanase and MMP-mediated articular cartilage<br />

degradation. Osteoarthritis Cartilage. 2010;18(3):279-288.<br />

18 Madsen S.H., Sondergaard B.C., Bay-Jensen A.C., and<br />

Karsdal M.A. Cartilage formation measured by a novel<br />

PIINP assay suggests that IGF-I does not stimulate but<br />

maintains cartilage formation ex vivo. Scand J Rheumatol.<br />

2009;38(3):222-226.


19 Brandt K.D. Animal models <strong>of</strong> osteoarthritis.<br />

Biorheology. 2002;39(1-2):221-235.<br />

20 Helminen H.J., Saamanen A.M., Salminen H., and<br />

Hyttinen M.M. Transgenic mouse models for studying the<br />

role <strong>of</strong> cartilage macromolecules in osteoarthritis.<br />

Rheumatology (Oxford). 2002;41(8):848-856.<br />

21 Glasson S.S. In vivo osteoarthritis target validation<br />

utilizing genetically-modified mice. Curr Drug Targets.<br />

2007;8(2):367-376.<br />

22 Manicourt D.H., Altman R.D., Williams J.M., Devogelaer<br />

J.P., Druetz-Van E.A., Lenz M.E., Pietryla D. et al.<br />

Treatment with calcitonin suppresses the responses <strong>of</strong><br />

<strong>bone</strong>, cartilage, and synovium in the early stages <strong>of</strong> canine<br />

experimental osteoarthritis and significantly reduces the<br />

severity <strong>of</strong> the cartilage lesions. Arthritis Rheum.<br />

1999;42(6):1159-1167.<br />

23 Behets C., Williams J.M., Chappard D., Devogelaer J.P.,<br />

and Manicourt D.H. Effects <strong>of</strong> calcitonin on <strong>subchondral</strong><br />

trabecular <strong>bone</strong> changes and on osteoarthritic cartilage<br />

lesions after acute anterior cruciate ligament deficiency. J<br />

Bone Miner Res. 2004;19(11):1821-1826.<br />

24 Bendele A.M., White S.L., and Hulman J.F. Osteoarthrosis<br />

in guinea pigs: histopathologic and scanning electron<br />

microscopic features. Lab Anim Sci. 1989;39(2):115-121.<br />

25 Jimenez P.A., Glasson S.S., Trubetskoy O.V., and Haimes<br />

H.B. Spontaneous osteoarthritis in Dunkin Hartley guinea<br />

pigs: histologic, radiologic, and biochemical changes. Lab<br />

Anim Sci. 1997;47(6):598-601.<br />

26 Sondergaard B.C. The Pharmacological Role <strong>of</strong> Calcitonin<br />

in Osteoarthritis. PhD thesis. 2010;41-41.<br />

CHAPTER 3: Overview <strong>of</strong> OA models<br />

49<br />

27 Goldring S.R. and Goldring M.B. The role <strong>of</strong> cytokines in<br />

cartilage matrix degeneration in osteoarthritis. Clin Orthop<br />

Relat Res. 2004;(427 Suppl):S27-S36.<br />

28 Goldring M.B. Update on the biology <strong>of</strong> the chondrocyte<br />

and new approaches to treating cartilage diseases. Best<br />

Pract Res Clin Rheumatol. 2006;20(5):1003-1025.<br />

29 Hui W., Rowan A.D., Richards C.D., and Cawston T.E.<br />

Oncostatin M in combination with tumor necrosis factor<br />

alpha induces cartilage damage and matrix<br />

metalloproteinase expression in vitro and in vivo. Arthritis<br />

Rheum. 2003;48(12):3404-3418.<br />

30 Kronenberg H.M. Developmental regulation <strong>of</strong> the<br />

growth plate. Nature. 2003;423(6937):332-336.<br />

31 van Osch G.J., Mandl E.W., Marijnissen W.J., van d., V,<br />

Verwoerd-Verhoef H.L., and Verhaar J.A. Growth factors<br />

in cartilage tissue engineering. Biorheology. 2002;39(1-<br />

2):215-220.<br />

32 Tyler J.A. Insulin-like growth factor 1 can decrease<br />

degradation and promote synthesis <strong>of</strong> proteoglycan in<br />

cartilage exposed to cytokines. Biochem J.<br />

1989;260(2):543-548.<br />

33 Verschure P.J., Van Noorden C.J., Van M.J., and Van den<br />

Berg W.B. Articular cartilage destruction in experimental<br />

inflammatory arthritis: insulin-like growth factor-1<br />

regulation <strong>of</strong> proteoglycan metabolism in chondrocytes.<br />

Histochem J. 1996;28(12):835-857.<br />

34 Wahl A.F. and Wallace P.M. Oncostatin M in the antiinflammatory<br />

response. Ann Rheum Dis. 2001;60 Suppl<br />

3:iii75-iii80.


Suzi Høgh Madsen, PhD thesis<br />

50


CHAPTER 4<br />

CHAPTER 4: PAPER I<br />

PAPER I<br />

___________________________________________________________________________<br />

Introductory remarks<br />

PAPER I validates a novel ex vivo model that allows investigation <strong>of</strong> cartilage and <strong>bone</strong> in one<br />

closed system. The development and validation <strong>of</strong> the novel ex vivo model, included selection and<br />

optimization processes that have not been included in PAPER I:<br />

Selection <strong>of</strong> femur head as the explant sample (the embryonic tibia was tested first)<br />

Sex <strong>of</strong> the mice<br />

Time <strong>of</strong> culture period<br />

Selection <strong>of</strong> conditioned medium<br />

51


Original Article<br />

Characterization <strong>of</strong> an Ex vivo Femoral<br />

Head Model Assessed by Markers <strong>of</strong><br />

Bone and Cartilage Turnover<br />

Suzi Hoegh Madsen 1 , Anne S<strong>of</strong>ie Goettrup 1 , Gedske Thomsen 1 ,<br />

Søren Tvorup Christensen 2 , Nikolaj Schultz 1 , Kim Henriksen 3 ,<br />

Anne-Christine Bay-Jensen 1 , and Morten Asser Karsdal 1<br />

Abstract<br />

Introduction<br />

Osteoarthritis (OA) is a degenerative joint disease leading<br />

to cartilage degradation and <strong>subchondral</strong> <strong>bone</strong> changes.<br />

Many factors contribute to the onset <strong>of</strong> OA, including both<br />

metabolic and biomechanical mechanisms, but it is still<br />

debated in which tissue the disease initiates. 1 In 1986, Dr.<br />

Radin and Dr. Rose 2 were the first to suggest that the<br />

<strong>subchondral</strong> <strong>bone</strong> plays an <strong>important</strong> role in the initiation<br />

and progression <strong>of</strong> OA. Physical characterizations <strong>of</strong> OA<br />

include cartilage degradation, <strong>subchondral</strong> <strong>bone</strong> sclerosis<br />

and thickening, and osteophyte formation. 3-5 In addition,<br />

trabecular <strong>bone</strong> in the <strong>subchondral</strong> region is thinned, and its<br />

elasticity is lost. 6-8<br />

An increasing line <strong>of</strong> evidence suggests that there is a<br />

strong interrelationship <strong>between</strong> <strong>subchondral</strong> <strong>bone</strong> and<br />

articular cartilage. Subchondral <strong>bone</strong> is separated from the<br />

articular cartilage by a thin layer <strong>of</strong> calcified cartilage. 9<br />

Cartilage<br />

2(3) 265 –278<br />

© The Author(s) 2011<br />

Reprints and permission:<br />

sagepub.com/journalsPermissions.nav<br />

DOI: 10.1177/1947603510383855<br />

http://cart.sagepub.com<br />

Objective: The pathophysiology <strong>of</strong> osteoarthritis involves the whole joint and is characterized by cartilage degradation<br />

and altered <strong>subchondral</strong> <strong>bone</strong> turnover. At present, there is a need for biological models that allow investigation <strong>of</strong> the<br />

<strong>interactions</strong> <strong>between</strong> the key cellular players in <strong>bone</strong>/cartilage: osteoblasts, osteoclasts, and chondrocytes. Methods: Femoral<br />

heads from 3-, 6-, 9-, and 12-week-old female mice were isolated and cultured for 10 days in serum-free media in the absence<br />

or presence <strong>of</strong> IGF-I (100 nM) (anabolic stimulation) or OSM (10 ng/mL) + TNF-α (20 ng/mL) (catabolic stimulation).<br />

Histology on femoral heads before and after culture was performed, and the growth plate size was examined to evaluate the effects<br />

on cell metabolism. The conditioned medium was examined for biochemical markers <strong>of</strong> <strong>bone</strong> and cartilage degradation/<br />

formation. Results: Each age group represented a unique system regarding the interest <strong>of</strong> <strong>bone</strong> or cartilage metabolism.<br />

Stimulation over 10 days with OSM + TNF-α resulted in depletion <strong>of</strong> proteoglycans from the cartilage surface in all ages.<br />

Furthermore, OSM + TNF-α decreased growth plate size, whereas IGF-I increased the size. Measurements from the<br />

conditioned media showed that OSM + TNF-α increased the number <strong>of</strong> osteoclasts by approximately 80% and induced<br />

<strong>bone</strong> and cartilage degradation by approximately 1200% and approximately 2600%, respectively. Stimulation with IGF-I<br />

decreased the osteoclast number and increased cartilage formation by approximately 30%. Conclusion: Biochemical markers<br />

and histology together showed that the catabolic stimulation induced degradation and the anabolic stimulation induced<br />

formation in the femoral heads. We propose that we have established an explant whole-tissue model for investigating cellcell<br />

<strong>interactions</strong>, reflecting parts <strong>of</strong> the processes in the pathogenesis <strong>of</strong> joint degenerative diseases.<br />

Keywords<br />

osteoarthritis, growth plate, ex vivo, femoral head, cytokines, IGF-I<br />

52<br />

How much do <strong>bone</strong> and its interaction with cartilage contribute<br />

to the causality <strong>of</strong> OA? It has been suggested that<br />

<strong>bone</strong> turnover increases in patients with OA. 3,4 In a canine<br />

model, <strong>subchondral</strong> <strong>bone</strong> loss was seen in the early OA and<br />

<strong>bone</strong> sclerosis in the late OA. 10,11 A murine OA model, an<br />

anterior cruciate ligament transaction (ACLT) model,<br />

showed that bisphosphonates, approved as antiresorptive<br />

1<br />

Cartilage Biology and Biomarkers R&D, Nordic Bioscience A/S, Herlev,<br />

Denmark<br />

2<br />

Department <strong>of</strong> Molecular Biology, University <strong>of</strong> Copenhagen,<br />

Copenhagen, Denmark<br />

3<br />

Bone Biology R&D, Nordic Bioscience A/S, Herlev, Denmark<br />

Corresponding Author:<br />

Suzi Hoegh Madsen, Nordic Bioscience A/S, Herlev Hovedgade 207, 2730<br />

Herlev, Denmark<br />

Email: shm@nordicbioscience.com


266 Cartilage 2(3)<br />

therapy for osteoporosis, effectively retarded the progression<br />

<strong>of</strong> cartilage degeneration and osteophyte formation,<br />

indicating the importance <strong>of</strong> <strong>bone</strong> remodeling in the pathogenesis<br />

<strong>of</strong> OA. 12 This and other studies suggest the existence<br />

<strong>of</strong> <strong>important</strong> <strong>interactions</strong> <strong>between</strong> <strong>bone</strong> and cartilage,<br />

which might be key events in OA pathogenesis. 13-17 However,<br />

human cartilage explants cultured in the presence <strong>of</strong> isolated<br />

osteoblasts from OA patients have shown increased<br />

proteoglycan degradation compared to cartilage cultured<br />

with osteoblasts from healthy patients. 13 Another group<br />

suggested that osteoblasts from sclerotic <strong>subchondral</strong> <strong>bone</strong><br />

could initiate chondrocyte hypertrophy. 14 The communication<br />

<strong>between</strong> osteoblasts and osteoclasts via PTH/RANKL<br />

in normal <strong>bone</strong> turnover, where PTH stimulates osteoblasts<br />

to release RANKL that in turn stimulates <strong>bone</strong> resorption<br />

via osteoclasts, is well acknowledged. 16 However, this<br />

communication via PTH/RANKL is altered in joint tissues<br />

from OA patients. 17 This lack <strong>of</strong> correlation suggests that<br />

the communication <strong>between</strong> the cells is very <strong>important</strong> for<br />

maintaining a healthy joint, and more attention should be<br />

directed toward investigating this communication.<br />

Bone and cartilage degradation can be measured by biochemical<br />

markers, such as fragments <strong>of</strong> C-terminal telopeptide<br />

<strong>of</strong> type I collagen (CTX-I), reflecting <strong>bone</strong> resorption, 18 and<br />

fragments from C-terminal telopeptide <strong>of</strong> type II collagen<br />

(CTX-II), reflecting cartilage degradation. 19,20 The turnover <strong>of</strong><br />

aggrecan, the predominating proteoglycan in cartilage, can be<br />

measured by the release <strong>of</strong> sulfated glycosaminoglycans<br />

(sGAGs) from the extracellular matrix (ECM). The formation<br />

<strong>of</strong> cartilage can be measured by the biomarker, PIINP, which<br />

is the collagen type II propeptide at the N-terminal. 21<br />

Biochemical markers for measuring biological processes,<br />

especially in OA, are valuable descriptive tools.<br />

A model that not only identifies the different changes in<br />

cartilage degradation but simultaneously changes adjacent<br />

<strong>bone</strong> may be useful for investigating <strong>interactions</strong> <strong>between</strong><br />

these 2 tissues. An in vitro model like this would allow us<br />

to investigate and evaluate potential effects <strong>of</strong> a given compound<br />

before in vivo experiments. Thus, such a model may<br />

be an <strong>important</strong> tool for finding potential joint disease–<br />

modifying drugs with “dualaction” targeting both <strong>bone</strong> and<br />

cartilage. Femoral heads from mice are small, enclosed<br />

compartments, which comprise both <strong>bone</strong> and cartilage.<br />

Thus, the interaction <strong>between</strong> osteoblasts, osteoclasts, and<br />

chondrocytes remains intact in the closed system when<br />

isolated from mice. The femoral heads are an open opportunity<br />

for investigating the signaling and communication<br />

<strong>between</strong> cells <strong>of</strong> <strong>bone</strong> and cartilage in near-normal conditions<br />

and under stimulation from a compound <strong>of</strong> interest.<br />

In this study, we developed and characterized a murine<br />

femoral head ex vivo model in which we were able to<br />

induce a catabolic or anabolic response. As controls, we<br />

chose oncostatin M (OSM) + tumor necrosis factor (TNF)-α<br />

and insulin-like growth factor (IGF)-I, which we have<br />

53<br />

previously established in other ex vivo models as catabolic<br />

and anabolic controls, respectively. 21,22 The idea for this<br />

model has emerged from a previously reported murine<br />

femoral head model, where only the cartilage compartment<br />

was cultured as part <strong>of</strong> investigating the enzymatic proteo-<br />

glycan degradation. 23<br />

Methods<br />

The Murine Femoral Head Explant Model<br />

Femoral heads from 3-, 6-, 9-, and 12-week-old NMRI<br />

(inbred) female mice (Charles River, Sulzfeld, Germany)<br />

were isolated by cutting at the femoral neck. The femoral<br />

heads were cultured in the media DMEM:F12 (Invitrogen,<br />

Taastrup, Denmark) + penicillin and streptomycin (P/S)<br />

(Lonza, Vallensbaek Strand, Denmark) + 10% serum<br />

(Invitrogen) for 2 hours to adjust to the isolation. The<br />

femoral heads were washed in serum-free DMEM:F12 +<br />

P/S 5 times and randomly placed in doublets in each well<br />

in a 24-well plate. Some femoral heads were saved directly<br />

in 4% formaldehyde (Merck, Hellerup, Denmark) to be<br />

able to evaluate the femoral heads at the point before culturing<br />

(T = 0). The rest <strong>of</strong> the femoral heads were cultured<br />

for 10 days in DMEM:F12 + P/S in the absence (nonstimulated<br />

control named W/O) or presence <strong>of</strong> the anabolic factor,<br />

IGF-I (100 nM) (Sigma-Aldrich, Copenhagen,<br />

Denmark), or catabolic cytokines, OSM (10 ng/mL)<br />

(Sigma-Aldrich) + TNF-α (20 ng/mL) (R&D Systems,<br />

Abingdon, UK). Each treatment was repeated 5 times. The<br />

conditioned media were changed and saved in –20 °C at<br />

days 4, 7, and 10, except for experiments with 12-week-old<br />

mice, which only were saved at day 10 (the conditioned<br />

media at day 10 comprises the total release from the whole<br />

culture period). Overall cell viability was monitored using<br />

the dye Alamar Blue (Invitrogen) at day 10. The femoral<br />

heads were washed 3 times in PBS (Lonza) and saved in<br />

4% formaldehyde. All culturing <strong>of</strong> femoral heads was at<br />

37°C and 5% CO 2 . For experiments with 3-, 6-, and 9-weekold<br />

mice, the conditioned media from days 4, 7, and 10<br />

were assessed by biochemical markers <strong>of</strong> <strong>bone</strong> and cartilage.<br />

All 3 days were accumulated into one graph, to represent<br />

the total release <strong>of</strong> the respective biochemical marker<br />

from the femoral heads, during the 10-day culture period.<br />

For the experiments with 12-week-old mice, the total<br />

releases were measured at day 10. The microscopic changes<br />

were assessed by histology.<br />

Biochemical Markers<br />

The biochemical marker CTX-I was used to detect type I collagen<br />

degradation fragments in the conditioned medium. The<br />

competitive ELISA used, RatLaps (IDS Ltd., Herlev,<br />

Denmark), quantified cathepsin K–mediated degradation


Madsen et al. 267<br />

products <strong>of</strong> type I collagen. The primary antibody was<br />

directed toward the neoepitope 1196 EKSQDGGR. 24 Serum<br />

Pre-clinical CartiLaps (IDS Ltd.) was used to quantify MMPmediated<br />

degradation products from type II collagen. The<br />

primary antibody was directed toward the neoepitope<br />

1230 EKGPDP. 24 PIINP (IDS Ltd.) was a biochemical marker for<br />

cartilage formation. The primary antibody was directed against<br />

an internal epitope <strong>of</strong> the N-terminal propeptides <strong>of</strong> collagen<br />

type II: 122 GPQGPAGEQGPRGDR 136 . 25 All 3 ELISAs were<br />

preformed accordingly to the manufacturer’s protocols.<br />

Measurement <strong>of</strong> Collagen Turnover<br />

The amino acid hydroxyproline was used as a measurement<br />

<strong>of</strong> total collagen release since it is a major component <strong>of</strong><br />

collagen. The 20 uL sample or standard (prepared from<br />

1-hydroxyproline diluted in 1 mM HCl) was diluted 5 times<br />

in 7.5 M HCl and hydrolyzed overnight (20 hours) at 110 °C.<br />

Samples were centrifuged for 2 minutes at 3000 g/min and<br />

then evaporated at 50 °C until samples were completely<br />

dry. Isopropanol/H 2 O (2:1) <strong>of</strong> 25 uL (Merck) was used to<br />

dissolve samples, and 10 uL <strong>of</strong> dissolved sample was transferred<br />

to a new plate, to which 20 uL isopropanol was<br />

added. This was oxidized by the addition <strong>of</strong> 10 uL <strong>of</strong> solution<br />

I (1 part <strong>of</strong> 24 M chloramines [Sigma-Aldrich] to 4<br />

parts <strong>of</strong> 1 M Na-acetate [Sigma-Aldrich], 0.33 M Na 3<br />

citrate [Merck] and 0.07 M citric acid [Merck] dissolved in<br />

isopropanol) incubated for 4 ± 1 minutes. There was 130 uL <strong>of</strong><br />

solution II (3 parts <strong>of</strong> 4.5 M 4-dimethylamino-benzaldehyde<br />

[Sigma-Aldrich] dissolved in 60% perchloric acid [Merck]<br />

to 13 parts <strong>of</strong> isopropanol) added and incubated at 60 °C for<br />

25 ± 5 minutes. The absorbance was measured at 558 nm<br />

on an ELISA reader.<br />

Quantification <strong>of</strong> Osteoclast Number<br />

TRAP was a quantitative measurement <strong>of</strong> osteoclast<br />

number. The reaction buffer (1.0 M sodium-acetate, 0.5%<br />

Triton X-100, 1 M NaCl, 10 mM EDTA [pH 5]) (Merck),<br />

50 mM ascorbic acid (Sigma-Aldrich), 0.2 M disodium<br />

tartrate (Merck), 82 mM 4-nitrophenylphosphate (Sigma-<br />

Aldrich), and milli = Q were mixed 2:1:1:1:3 into a final<br />

TRAP solution buffer. Sample and final TRAP solution<br />

buffer was mixed (1:4) and incubated for 1 hour at 37 °C.<br />

There was 0.3 M NaOH (Merck) added to stop the color<br />

reaction, and the absorbance was measured at 405 nm with<br />

650 nm as reference.<br />

Quantification <strong>of</strong> Proteoglycans by Measurement <strong>of</strong><br />

Sulfated Glycoaminoglycans<br />

Proteoglycans were used to quantify cartilage turnover by<br />

measuring sulfated glycosaminoglycans. An assay using the<br />

dye 1.9-dimethylmehylene blue chloride (Sigma-Aldrich)<br />

54<br />

in a solution <strong>of</strong> sodium formate (Sigma-Aldrich) and formic<br />

acid (Merck) was used. A standard curve was prepared<br />

with chondroitin sulfate (Sigma-Aldrich) ranging from 0 to<br />

100 ng/mL. The 40 uL standard or sample was transferred<br />

to a microtiter plate, followed by 250 uL 38.5 uM<br />

1.9-dimethylmethylene blue chloride. The absorbance was<br />

immediately read at 650 nm on an ELISA reader to avoid<br />

precipitation.<br />

Histological Analysis<br />

Tissue preparation. After the culture period, femoral heads<br />

were decalcified in 15% EDTA at room temperature for<br />

approximately 1 week. Femoral heads were embedded separately<br />

in paraffin blocks and cut into 5-µm-thick sections.<br />

Excessive paraffin was removed by placing sections at 60 °C<br />

for 1 hour and dried at 37 °C overnight before staining.<br />

Safranin O and fast green staining. To visualize <strong>bone</strong> and<br />

cartilage, safranin O (Sigma-Aldrich) stained proteoglycans<br />

red, and fast green (Sigma-Aldrich) stained collagens<br />

green. Cartilage was stained red due to the excess <strong>of</strong> proteoglycans<br />

compared to collagens. Sections (5 µm) were<br />

stained according to Bay-Jensen et al. 26 Digital histographs<br />

were taken using an Olympus BX60F-3 microscope and an<br />

Olympus DP71 camera (Tokyo, Japan).<br />

Annotations <strong>of</strong> the femoral head to measure size <strong>of</strong> the<br />

growth plate. Digital histographs <strong>of</strong> femoral heads from<br />

12-week-old mice were evaluated by standardized annotations,<br />

ensuring anatomical correspondence. A line connects<br />

the black dots where the cartilage starts on the femoral<br />

neck. The center <strong>of</strong> this line is perpendicularly connected to<br />

the cartilage surface, and 2 lines are placed at an angle <strong>of</strong><br />

30° from this perpendicular line. The lengths <strong>of</strong> the section<br />

<strong>of</strong> the 3 lines where they cross the growth plate are calculated<br />

to find the average length <strong>of</strong> the growth plate. The<br />

growth plate was evaluated in 4 different femoral heads<br />

(from 4 different experiments) from each treatment.<br />

Statistical Analysis<br />

Results are shown as mean ± standard error <strong>of</strong> the mean<br />

(SEM). Differences <strong>between</strong> mean values were compared by<br />

the Student t test for unpaired observations, assuming normal<br />

distribution where 5 replicates were used. Differences were<br />

considered statistically significant if P < 0.05.<br />

Results<br />

Femoral Heads Exhibit Different Bone and Cartilage<br />

Morphology during Development<br />

Each individual age has a unique <strong>bone</strong> and cartilage morphology.<br />

In 3-week-old mice, the cartilage compartment is<br />

dominating (red stain) compared to the <strong>bone</strong> compartment


268 Cartilage 2(3)<br />

Figure 1. Characterization <strong>of</strong> the development in murine femoral heads. Sections from femoral heads from 3-, 6-, 9-, and 12-week-old<br />

mice were stained with safranin O and fast green, which color the proteoglycans red and the collagens green. The black bars represent<br />

250 µM (A-D) and 100 µM (E-L). The numbers represent growth plate (1), ligamentum teres femoris (2), endochondral ossification (3),<br />

immature <strong>bone</strong> embedded with calcified cartilage (4), prehypertrophic and hypertrophic chondrocytes (5), calcified cartilage (6), woven<br />

<strong>bone</strong> (unmineralized) (7), lamellar <strong>bone</strong> (mineralized) (8), secondary ossification center (9), and articular cartilage (10). The black squares<br />

in the whole femoral heads (A-D) represent the magnification <strong>of</strong> either the cartilage compartment (E-H) or the <strong>bone</strong> compartment<br />

(I-L), which is divided in columns after age.<br />

(green stain); however, the cartilage is not fully materialized<br />

(Fig. 1A). The cartilage-<strong>bone</strong> ratio changes over time<br />

due to the development <strong>of</strong> the secondary ossification center<br />

at the age <strong>of</strong> 9 weeks (Fig. 1C). The growth plate is visible<br />

as a thick line <strong>of</strong> dense red staining separating <strong>bone</strong> from<br />

cartilage in the 3-week-old mouse, which decreases concurrently<br />

with increasing age (Fig. 1A-D). In femoral heads<br />

<strong>of</strong> 3-week-old mice, the cartilage predominantly consists <strong>of</strong><br />

proliferating, prehypertrophic, and hypertrophic chondrocytes<br />

(Fig. 1E), whereas the 6- and 9-week-old mice have<br />

less proliferating chondrocytes, which are located in the<br />

reduced growth plate (Fig. 1F and G). Additionally, the<br />

extracellular matrix <strong>of</strong> the cartilage compartment in 3- to<br />

9-week-old mice is calcified (Fig. 1E-G), preparing itself<br />

for the secondary ossification center, whereas the cartilage<br />

compartment is mainly articular cartilage (noncalcified) at<br />

12 weeks <strong>of</strong> age (Fig. 1H). The <strong>bone</strong> compartment in the<br />

3-week-old mice consists <strong>of</strong> immature <strong>bone</strong> embedded with<br />

calcified cartilage (Fig. 1I). As the mice get older, the calcified<br />

cartilage is replaced by unmineralized <strong>bone</strong> (normal<br />

endochondral ossification process), resulting in woven<br />

<strong>bone</strong> with a high proportion <strong>of</strong> osteocytes (Fig. 1J and K).<br />

The mechanically weak woven <strong>bone</strong> is replaced by more<br />

55<br />

resilient lamellar <strong>bone</strong> (mechanically strong) with a low<br />

proportion <strong>of</strong> osteocytes when the mice reach the age <strong>of</strong> 12<br />

weeks (Fig. 1L). Overall, these results suggest that the different<br />

ages represent different developmental stages, as<br />

expected, especially with regards to ratio and quality <strong>of</strong><br />

<strong>bone</strong> and cartilage, and thus, these can individually be used<br />

in our ex vivo model for specific purposes.<br />

Anabolic and Catabolic Stimulations Induce<br />

Morphological Changes<br />

The viability <strong>of</strong> the cells <strong>of</strong> the cultured femoral heads is<br />

not impaired by the 10 days <strong>of</strong> anabolic or catabolic stimulation<br />

(data not shown). Femoral heads stimulated with<br />

OSM + TNF-α show a major loss <strong>of</strong> proteoglycans from the<br />

cartilage compartment at the age <strong>of</strong> 3 weeks (Fig. 2F) compared<br />

to the nonstimulated control (W/O) (Fig. 2A). In<br />

9-week-old femoral heads, OSM + TNF-α stimulation<br />

increases the loss <strong>of</strong> proteoglycans from the cartilage surfaces<br />

(Fig. 2H) compared to the nonstimulated controls<br />

(W/O) (Fig. 2C). However, in femoral heads from 6-weekold<br />

mice, both nonstimulated (Fig. 2B) and OSM + TNFα–stimulated<br />

(Fig. 2G) femoral heads have lost proteoglycans


Madsen et al. 269<br />

Figure 2. Microscopic changes after catabolic and anabolic stimulation. Sections from femoral heads from 3-, 6-, 9-, and 12-week-old<br />

mice were stained with safranin O (proteoglycans) and fast green (collagens) after culture for 10 days in the absence (W/O) or presence<br />

<strong>of</strong> IGF-I (anabolic) and OSM + TNF-α (catabolic) stimulation. The black bars represent 250 µM (A-D, F-I, K-N) and 20 µM (E, J, O). The<br />

black squares in the 12-week-old femoral heads (D, I, N) represent the magnification <strong>of</strong> the cartilage surface (E, J, O), which is divided<br />

in rows after treatment.<br />

from the cartilage surface. In 12-week-old femoral heads,<br />

the proteoglycans seem to be almost depleted from the cartilage<br />

compartment in the nonstimulated (Fig. 2D) and the<br />

OSM + TNF-α–stimulated (Fig. 2I) explants. Anabolic<br />

stimulation with IGF-I protects against the proteoglycan<br />

loss seen in W/O (Fig. 2A-D and K-N). Additionally, in<br />

12-week-old mice, anabolic stimulation shows proteoglycan<br />

staining around the chondrocytes (Fig. 2O) compared to the<br />

W/O (Fig. 2E). Next, we measured the size <strong>of</strong> the growth<br />

plate to investigate whether morphological changes could<br />

be induced (Fig. 3A). The size <strong>of</strong> the growth plate in IGF-<br />

I–stimulated 12-week-old femoral heads is increased by<br />

approximately 48% (P < 0.001) compared to W/O, whereas<br />

OSM + TNF-α stimulation decreases the growth plate size<br />

by approximately 34% (P < 0.001) compared to W/O (Fig.<br />

3B). This indicates that we are able to induce alteration to<br />

the femoral heads ex vivo with catabolic (degenerative) or<br />

anabolic (generative) factors.<br />

Catabolic Stimulation Increases Collagen and<br />

Proteoglycan Turnover<br />

Levels <strong>of</strong> hydroxyproline, an indicator <strong>of</strong> total collagen<br />

turnover, in the conditioned medium indicate that stimulation<br />

by OSM + TNF-α increases the total release <strong>of</strong> collagens by<br />

approximately 49% (P < 0.05) to approximately 190%<br />

(P < 0.001) in femoral heads aged 6, 9, and 12 weeks, but not in<br />

femoral heads from 3-week-old mice (Fig. 4A). The longitudinal<br />

effects from the different stimulations are shown in<br />

Table 1. OSM + TNF-α increases the release <strong>of</strong> collagens<br />

56<br />

Figure 3. Quantification <strong>of</strong> growth plate zone in femoral heads.<br />

Sections from femoral heads from 12-week-old mice were stained<br />

with safranin O (proteoglycans) and fast green (collagens) after<br />

culture for 10 days in the absence (W/O) or presence <strong>of</strong> IGF-I<br />

(anabolic) and OSM + TNF-α (catabolic) stimulation. The growth<br />

plate size was measured in one femoral head per treatment from 4<br />

different experiments using the annotation system (see Methods).<br />

The growth plate thickness (µm) is calculated by the average <strong>of</strong><br />

the black lines in the growth plate (A). The catabolic stimulation<br />

decreased the growth plate size, whereas the anabolic stimulation<br />

increased the size <strong>of</strong> the growth plate (B). ***P < 0.001.<br />

over time in 6- and 9-week-old mice. In femoral heads from<br />

12-week-old mice, the anabolic stimulation also increases<br />

total collagen release by approximately 49% (P < 0.01)<br />

compared to W/O (Fig. 4A). OSM + TNF-α stimulation<br />

increases the total release <strong>of</strong> sulfated glycosaminoglycans<br />

(sGAG) to the conditioned media by approximately 22%<br />

(P < 0.05) to approximately 40% (P < 0.01), except from<br />

the experiment with 6-week-old mice (Fig. 4B); however,<br />

similar trends were observed.


270 Cartilage 2(3)<br />

Figure 4. The catabolic control induces a release <strong>of</strong> collagens and proteoglycans. The experiments were assessed by the collagen<br />

degradation marker, hydroxyproline (A), and the proteoglycan turnover marker, sGAG (B). Each graph represents the accumulated<br />

measurements from days 4, 7, and 10 except from 12-week-old mice (only day 10). Cytokine stimulation increases the collagen release<br />

in experiments with 6-, 9-, and 12-week-old mice (A). Cytokine stimulation also increases proteoglycan release in experiments with 3-,<br />

9-, and 12-week-old mice (B). Asterisks indicate statistical significance: *P < 0.05; **P < 0.01; ***P < 0.001.<br />

57


Madsen et al. 271<br />

Table 1. Biochemical Markers Measured from the Conditioned Medium at Days 4, 7, and 10 in the Different Age Groups<br />

Age <strong>of</strong> Culture Stimulated<br />

Mice, wk Day with HP, µg/mL sGAG, µg/mL TRAP CTX-I, ng/mL PIINP, ng/mL CTX-II, pg/mL<br />

W/O 5.22 ± 0.81 113.12 ± 3.46 0.47 ± 0.06 2.97 ± 0.82 93.61 ± 4.65 48.69 ± 3.73<br />

4 O + T 6.21 ± 1.22 143.84 ± 8.45** 0.81 ± 0.12* 3.23 ± 0.27 54.92 ± 9.58** 74.96 ± 19.19<br />

IGF 5.16 ± 0.59 92.61 ± 10.11* 0.63 ± 0.06 3.79 ± 0.84 100 ± 0.00 26.10 ± 3.87**<br />

W/O 2.59 ± 0.46 49.51 ± 8.51 0.27 ± 0.02 4.61 ± 0.54 68.78 ± 5.55 6.30 ± 1.40<br />

3 7 O + T 2.41 ± 0.35 87.18 ± 5.04** 0.33 ± 0.01* 11.72 ± 3.54 40.83 ± 3.02** 17.44 ± 3.83*<br />

IGF 4.38 ± 0.46* 58.96 ± 4.34 0.26 ± 0.01 4.49 ± 0.39 74.38 ± 1.76 15.28 ± 1.86**<br />

W/O 3.56 ± 0.53 70.74 ± 4.85 0.22 ± 0.001 2.55 ± 0.56 46.01 ± 3.05 0.48 ± 0.38<br />

10 O + T 3.09 ± 0.29 83.18 ± 9.70** 0.25 ± 0.01** 3.62 ± 0.47 28.95 ± 2.11** 7.90 ± 1.93**<br />

IGF 4.53 ± 0.55 89.71 ± 3.88* 0.21 ± 0.01 2.26 ± 0.17 69.24 ± 1.38*** 8.51 ± 1.50**<br />

W/O 1.34 ± 0.16 40.52 ± 5.05 0.29 ± 0.03 3.21 ± 0.39 54.55 ± 6.48 10.70 ± 0.86<br />

4 O + T 1.69 ± 0.11 50.53 ± 0.68 0.36 ± 0.03 5.29 ± 0.76* 64.01 ± 4.38 29.66 ± 4.14**<br />

IGF 1.27 ± 0.09 24.16 ± 3.10 0.21 ± 0.01* 4.26 ± 0.16* 82.18 ± 6.23* 8.38 ± 2.30<br />

W/O 1.48 ± 0.28 24.22 ± 4.24 0.24 ± 0.01 3.11 ± 0.65 74.48 ± 2.48 16.26 ± 1.68<br />

6 7 O + T 1.57 ± 0.13 20.86 ± 1.01 0.46 ± 0.05** 13.77 ± 4.18* 59.43 ± 3.58** 139.18 ± 27.05**<br />

IGF 1.77 ± 0.06 24.86 ± 2.79 0.18 ± 0.002*** 4.21 ± 0.47 64.86 ± 5.31 14.81 ± 2.50<br />

W/O 1.53 ± 1.02 6.42 ± 1.32 0.19 ± 0.01 2.08 ± 0.14 56.22 ± 1.54 12.94 ± 1.70<br />

10 O + T 2.37 ± 0.38 10.81 ± 1.00 0.45 ± 0.06** 6.02 ± 1.07** 48.12 ± 4.02 160.84 ± 45.55*<br />

IGF 2.02 ± 0.45 10.35 ± 0.51 0.19 ± 0.02 4.58 ± 0.29*** 44.61 ± 5.05 14.47 ± 2.19<br />

W/O 2.26 ± 0.40 25.56 ± 1.35 0.33 ± 0.03 10.82 ± 0.84 65.45 ± 3.70 68.75 ± 11.89<br />

4 O + T 1.61 ± 0.38 28.26 ± 2.72 0.36 ± 0.04 14.46 ± 2.72 54.89 ± 3.22 59.61 ± 9.14<br />

IGF 2.05 ± 0.37 22.08 ± 2.46 0.24 ± 0.004** 4.80 ± 0.29*** 78.57 ± 2.69* 36.53 ± 8.24<br />

W/O 1.49 ± 0.35 9.70 ± 1.93 0.36 ± 0.04 14.86 ± 2.50 53.16 ± 4.19 32.18 ± 3.13<br />

9 7 O + T 3.01 ± 0.88 15.95 ± 1.56* 0.48 ± 0.06 18.71 ± 1.98 46.87 ± 2.82 309.83 ± 34.52***<br />

IGF 1.49 ± 0.27 11.39 ± 2.18 0.23 ± 0.01** 14.87 ± 2.68 66.78 ± 2.62* 18.63 ± 5.09<br />

12 a<br />

W/O 2.34 ± 0.41 14.12 ± 2.34 0.30 ± 0.02 9.09 ± 1.07 29.98 ± 3.36 30.86 ± 6.86<br />

10 O + T 6.76 ± 0.13*** 20.10 ± 0.91* 0.55 ± 0.07** 24.13 ± 3.33** 33.73 ± 2.81 361.52 ± 2.09***<br />

IGF 3.05 ± 0.81 13.32 ± 1.35 0.23 ± 0.03 6.25 ± 0.49* 33.09 ± 1.78 37.77 ± 13.16<br />

W/O 4.79 ± 0.53 19.74 ± 1.31 0.19 ± 0.008 27.84 ± 8.69 20.31 ± 3.61 62.88 ± 28.45<br />

10 O + T 13.89 ± 1.59*** 24.00 ± 1.39* 0.30 ± 0.037* 341.39 ± 78.25*** 0.80 ± 0.80* 1714.80 ± 379.31**<br />

IGF 7.15 ± 0.43** 23.67 ± 1.11 0.17 ± 0.011 42.60 ± 16.09 67.78 ± 0.73*** 54.07 ± 22.04<br />

Note: Values are significantly different from W/O.<br />

a The biochemical markers released from 12-week-old mice at day 10 represent total release from the entire culture period.<br />

*P < 0.05. **P < 0.01. ***P < 0.001.<br />

OSM + TNF-α Increases the Osteoclast Number<br />

and Bone Resorption<br />

Measurements <strong>of</strong> the conditioned media show that stimulation<br />

with OSM + TNF-α increases the tartrate-resistant<br />

acid phosphatase (TRAP) activity for all the age groups in<br />

a range from approximately 41% (P < 0.05) to approximately<br />

78% (P < 0.01) compared with their respective<br />

W/O, indicating increased osteoclast numbers (Fig. 5A).<br />

OSM + TNF-α increases the osteoclast number over time<br />

in 6- and 9-week-old mice. However, the osteoclast number<br />

decreases over time in 3-week-old mice (Table 1).<br />

Conversely to OSM + TNF-α stimulation, IGF-I stimulation<br />

shows approximately 17% (P < 0.05) to approximately<br />

29% (P < 0.01) decrease in the osteoclast number<br />

58<br />

in 6- and 9-week-old mice (Fig. 5A), which results from<br />

constant low values measured at all 3 days (Table 1). The<br />

collagen type I resorption measurements reflect the<br />

respective osteoclast number during catabolic stimulation.<br />

The resorption increases approximately 65% (P <<br />

0.05) to approximately 1200% (P < 0.001) in the presence<br />

<strong>of</strong> OSM + TNF-α stimulation compared to W/O (Fig. 5B).<br />

However, in 6-week-old mice, the collagen type I resorption<br />

measurements do not reflect the TRAP activity when<br />

stimulated with IGF-I. In 3- and 6-week-old mice, the<br />

resorption <strong>of</strong> collagen type I peaks at day 7, whereas it<br />

peaks at or after day 10 in 9-week-old mice (Table 1).<br />

These measurements show that it is possible to measure<br />

osteoclast number and activity via biochemical markers in<br />

our ex vivo model.


272 Cartilage 2(3)<br />

Figure 5. Cytokines increase the osteoclast number and resorption <strong>of</strong> <strong>bone</strong>. The conditioned media from the 4 experiments with 4<br />

individual age groups were assessed by the osteoclast number measured by TRAP activity (A) and the collagen type I resorption marker,<br />

CTX-I (B). Each graph represents the accumulated measurements from days 4, 7, and 10 except from 12-week-old mice (only day 10).<br />

Catabolic stimulation increases the osteoclast number in all 4 experiments, whereas the anabolic stimulation decreases the osteoclast<br />

number in the experiments with 6- and 9-week-old mice (A). Cytokines (O + T) also increase the total collagen type I resorption in the<br />

experiments with 6-, 9-, and 12-week-old mice (B). *P < 0.05. **P < 0.01. ***P < 0.001.<br />

59


Madsen et al. 273<br />

IGF-I and OSM + TNF-α Stimulation Mediate<br />

Collagen Type II Formation and Degradation,<br />

Respectively<br />

Collagen type II formation, measured by the PIINP released<br />

into the conditioned medium, decreases over time (Table 1).<br />

However, the total collagen type II formation increases by<br />

approximately 22% (P < 0.05), approximately 21% (P <<br />

0.05), and approximately 230% (P < 0.001) in anabolic<br />

stimulated (IGF-I) femoral heads from 3-, 9-, and 12-weekold<br />

mice, respectively, when compared to W/O (Fig. 6A).<br />

OSM + TNF-α decreases collagen type II formation by<br />

approximately 38% (P < 0.01) and approximately 96%<br />

(P < 0.05) compared to W/O in 3- and 12-week-old mice,<br />

respectively (Fig. 6A).<br />

Stimulation with OSM + TNF-α increases the degradation<br />

<strong>of</strong> collagen type II by approximately 450% (P < 0.001)<br />

to approximately 2600% (P < 0.01) in femoral heads from<br />

mice aged 6, 9, and 12 weeks, while femoral heads from<br />

3-week-old mice show only a nonsignificant increase<br />

(~80%) compared with W/O (Fig. 6B). The longitudinal<br />

release pattern <strong>of</strong> CTX-II shows that OSM + TNF-α<br />

increases the collagen type II degradation over time in 6-<br />

and 9-week-old mice (Table 1). In 3-week-old mice, the<br />

release <strong>of</strong> CTX-II decreases over time (Table 1). These<br />

results indicate that we are able to induce cartilage formation<br />

and degradation in our ex vivo model.<br />

Discussion<br />

OA is a complex disease <strong>of</strong> the entire joint affecting both<br />

<strong>bone</strong> and cartilage. 12,27 Currently, there are no effective<br />

disease-modifying OA drug (DMOAD) treatments. 28 We<br />

have developed an ex vivo murine femoral head model,<br />

comprising both <strong>bone</strong> and cartilage in one closed compartment<br />

system that allows us to identify changes in <strong>bone</strong> and<br />

cartilage simultaneously. This model allows <strong>interactions</strong><br />

<strong>between</strong> cartilage and <strong>bone</strong> cells—chondrocytes, osteoblasts,<br />

and osteoclasts—in a manner resembling their in situ<br />

conditions and microenvironment. However, the availability<br />

for humoral communication, due to the direct contact<br />

<strong>between</strong> the culture media and the different tissues, far<br />

exceeds what is possible in vivo. Furthermore, the ex vivo<br />

model does not mimic the biomechanical communication<br />

<strong>between</strong> <strong>bone</strong> and cartilage that is part <strong>of</strong> both normal<br />

physiology and pathophysiology. Nevertheless, the model<br />

is an <strong>important</strong> fundamental tool, allowing us to find preliminary<br />

results for potential therapies targeting one or both<br />

tissues. These preliminary results could aid the selection <strong>of</strong><br />

which future in vivo experiments, concerning joint diseases,<br />

should be investigated.<br />

We were able to induce catabolic and anabolic responses by<br />

stimulation with catabolic and anabolic factors, respectively.<br />

60<br />

Catabolic stimulation resulted in increased <strong>bone</strong> resorption<br />

and degradation <strong>of</strong> cartilage, whereas anabolic stimulation<br />

had a protective effect against proteoglycan loss and<br />

resulted in increased cartilage formation.<br />

Even though the different developmental stages <strong>of</strong><br />

endochondral ossification, including that <strong>of</strong> the femoral<br />

head, are known from the literature already, 29 it was <strong>important</strong><br />

to determine the individual stages in our particular<br />

murine model for comparison with the available literature.<br />

The microscopic evaluation <strong>of</strong> femoral heads from mice<br />

aged 3 to 12 weeks revealed unique <strong>bone</strong> and cartilage<br />

morphologies.<br />

In 3-week-old mice, the cartilage had a high metabolic<br />

rate due to the ongoing endochondral ossification process.<br />

The cartilage compartment was dominating compared to<br />

the <strong>bone</strong> compartment and mainly consisted <strong>of</strong> proliferating<br />

and hypertrophic chondrocytes, which included a large<br />

and active growth plate. Hypertrophic chondrocytes are a<br />

hallmark <strong>of</strong> early OA, 30 so this murine age group is highly<br />

relevant for preliminary studies, for finding potential<br />

DMOADs, by investigating the complicated nature <strong>of</strong> this<br />

phenotype. This age group is also relevant for investigating<br />

the metabolism <strong>of</strong> very active cartilage in vitro.<br />

In mice aged 6 and 9 weeks, the ratio <strong>of</strong> <strong>bone</strong> to cartilage<br />

was more even, and an emerging secondary ossification<br />

center had started to penetrate the cartilage compartment<br />

in 9-week-old mice. The <strong>bone</strong> compartment was deposited<br />

as woven <strong>bone</strong> with a high proportion <strong>of</strong> osteocytes. Woven<br />

<strong>bone</strong> forms quickly when osteoblasts produce osteoid,<br />

which occurs initially in all fetal <strong>bone</strong>s but is weak due to<br />

a disorganized collagen structure. The growth plate was<br />

reduced by more than 70%, separating the woven <strong>bone</strong><br />

from the indefinable mix <strong>of</strong> prehypertrophic and hypertrophic<br />

chondrocytes (in calcified cartilage) in an evenlooking<br />

arc. These 2 age groups are unique for investigating<br />

the metabolism <strong>of</strong> hypertrophic chondrocytes in the presence<br />

<strong>of</strong> woven <strong>bone</strong> (comprised <strong>of</strong> osteoclasts, osteoblasts,<br />

and osteocytes) or for studying the initial development <strong>of</strong><br />

the secondary ossification center.<br />

In 12-week-old mice, the <strong>bone</strong> compartment had become<br />

larger than the cartilage compartment, in direct contrast to<br />

the situation with 3-week-old mice. The calcified cartilage<br />

had been replaced by woven <strong>bone</strong> during the secondary<br />

ossification, which was enclosed by noncalcified cartilage,<br />

resembling mature articular cartilage. The woven <strong>bone</strong><br />

from the endochondral ossification had been replaced by<br />

the stronger and more resilient lamellar <strong>bone</strong>, which was<br />

highly organized in concentric collagen sheets with a low<br />

proportion <strong>of</strong> osteocytes. A small growth plate was still<br />

present and separated the endochondral ossification (lamellar<br />

<strong>bone</strong>) from the secondary ossification (woven <strong>bone</strong>).<br />

This older age group is unique for investigating mature<br />

<strong>bone</strong> and <strong>subchondral</strong> <strong>bone</strong> metabolism in the presence <strong>of</strong>


274 Cartilage 2(3)<br />

Figure 6. The anabolic and catabolic controls induce cartilage formation and degradation, respectively. The experiments were assessed<br />

by the collagen type II formation marker, PIINP (A), and the degradation marker, CTX-II (B). Each graph represents the accumulated<br />

measurements from days 4, 7, and 10 except from 12-week-old mice (only day 10). IGF-I induces formation in the experiments with 3-,<br />

9-, and 12-week-old mice (A). Cytokines (O + T) increased the total cartilage degradation in experiments with 6-, 9-, and 12-week-old<br />

mice (B). *P < 0.05. **P < 0.01. ***P < 0.001.<br />

61


Madsen et al. 275<br />

a reduced quantity <strong>of</strong> mature articular cartilage. Importantly,<br />

12-week-old mice resemble a human joint compared to the<br />

other age groups. Furthermore, the model system enables<br />

us to induce anabolic or catabolic responses, which mimic<br />

parts <strong>of</strong> the processes from different <strong>bone</strong> and cartilage<br />

diseases in an adult individual.<br />

The microscopic evaluation <strong>of</strong> femoral heads cultured<br />

for 10 days in the absence or presence <strong>of</strong> IGF-I (anabolic)<br />

or OSM + TNF-α (catabolic) stimulation showed that<br />

femoral heads stimulated with OSM + TNF-α lost more<br />

proteoglycans from the cartilage compartment than the<br />

W/O at almost all age groups. Nonstimulated femoral<br />

heads from 6- and 12-week-old mice also lost proteoglycans<br />

from the cartilage, as seen in the OSM + TNF-α<br />

stimulation. This suggests that the proteoglycan loss may<br />

be a spontaneous effect from the culturing rather than a<br />

catabolic effect from the OSM + TNF-α stimulation. The<br />

femoral heads stimulated with the anabolic factor (IGF-I)<br />

showed that this loss and depletion <strong>of</strong> proteoglycans during<br />

culture were protected compared to the W/O for all 4 age<br />

groups. Furthermore, in 12-week-old mice, anabolic stimulation<br />

showed far more proteoglycan staining around the<br />

chondrocytes compared to the W/O, indicating a regenerative<br />

effect <strong>of</strong> IGF-I, as previously observed in bovine cartilage<br />

explants. 31 We also showed that IGF-I stimulation in<br />

12-week-old mice increased the growth plate size significantly,<br />

whereas the OSM + TNF-α stimulation significantly<br />

decreased the size. These findings corroborate previous<br />

findings in the literature; IGF-I is very <strong>important</strong> for the<br />

growth plate during <strong>bone</strong> growth as it increases proliferation<br />

<strong>of</strong> resting and proliferative chondrocytes, and OSM +<br />

TNF-α has, on the contrary, shown to decrease the rate <strong>of</strong><br />

endochondral <strong>bone</strong> growth. 32-34<br />

Biochemical markers are valuable, longitudinal, and<br />

dynamic tools, which in combination with histology can<br />

assess the systematic effects <strong>of</strong> specific compounds. The<br />

histology shows the end result <strong>of</strong> the effect <strong>of</strong> a specific<br />

compound on the femoral head, whereas biochemical<br />

markers reveal which protein processes are up-regulated<br />

and down-regulated and in which order they are released.<br />

We have chosen bar graphs to present the total release <strong>of</strong><br />

the biochemical markers to the conditioned media as accumulated<br />

data from all media changes. Table 1 presents all<br />

the details for the individual days.<br />

Hydroxyproline measurements showed that the release<br />

<strong>of</strong> collagen fragments increased in the presence <strong>of</strong> OSM +<br />

TNF-α stimulation, except from 3 weeks <strong>of</strong> age. However,<br />

IGF-I also increased the release <strong>of</strong> collagens in 12-weekold<br />

mice. If this collagen release results from increased<br />

degradation, increased turnover or lack <strong>of</strong> incorporation<br />

into the extracellular matrix after synthesis may be an irrelevant<br />

discussion for our use. We can use the results <strong>of</strong> “collagen<br />

release” in combination with other biochemical<br />

62<br />

markers <strong>of</strong> collagens in our model. We can furthermore not<br />

exclude the fact that some portion <strong>of</strong> the hydroxyproline<br />

measured is released from the ligamentum teres.<br />

The release <strong>of</strong> proteoglycans from the femoral heads<br />

decreased proportionally with the increasing age <strong>of</strong> the<br />

mice, indicating that the release <strong>of</strong> proteoglycans mainly<br />

comes from the cartilage compartment and not the <strong>bone</strong>.<br />

However, a limitation <strong>of</strong> the current study is that we were<br />

not able to analyze the specific proteolytic mechanisms<br />

responsible for sGAG release. Future studies should thus<br />

aim to determine the longitudinal patterns <strong>of</strong> MMP- and<br />

ADAMTS-mediated aggrecan degradation in the femur<br />

head model and compare to other in vitro and in vivo models<br />

<strong>of</strong> <strong>bone</strong> and cartilage diseases. To evaluate the results<br />

further, we then measured more protein-specific biochemical<br />

markers.<br />

We found that OSM + TNF-α stimulation for 10 days<br />

increased the number <strong>of</strong> osteoclasts and increased the<br />

resorption <strong>of</strong> collagen type I. Even though the total release<br />

<strong>of</strong> TRAP increased in the presence <strong>of</strong> OSM + TNF-α (in<br />

3-week-old mice), the osteoclast number decreased over<br />

time for both nonstimulated and stimulated treatments.<br />

However, the TRAP concentrations at day 4 were higher<br />

than the concentration at day 10 for the other age groups.<br />

This incoherence might be explained by a peak at day 4 for<br />

the 3-week-old mice, in which we did not find a large<br />

amount <strong>of</strong> <strong>bone</strong>. The increased resorption seen in this<br />

model is consistent with the literature, which reports that<br />

<strong>subchondral</strong> <strong>bone</strong> was lost in the early stage <strong>of</strong> OA,<br />

whereas <strong>bone</strong> sclerosis was present at the late stage <strong>of</strong><br />

OA. 10,11 Furthermore, TNF-α can induce osteoclast differentiation<br />

in vitro through a mechanism independent <strong>of</strong><br />

RANKL. 35,36 Additionally, in murine osteoblastic MC3T3-E1<br />

cells, M-CSF expression is constitutive and can be further<br />

increased by TNF-α. 37-39 OSM can, in vitro, induce the<br />

formation <strong>of</strong> osteoclasts, leading to <strong>bone</strong> resorption.<br />

However, OSM can also influence differentiation and proliferation<br />

<strong>of</strong> osteoblasts, leading to <strong>bone</strong> formation. 40-42<br />

The anabolic stimulation with IGF-I generally showed,<br />

in all the age groups, a tendency to decrease the number <strong>of</strong><br />

osteoclasts, indicating a protective effect on the <strong>bone</strong> by<br />

reducing <strong>bone</strong> resorption. However, the collagen type I<br />

resorption did not decrease but seemed to be more or less<br />

unaffected by the anabolic stimulation. Interestingly, collagen<br />

type I resorption data did not reflect the anabolic<br />

TRAP data from mice aged 6 weeks. The femoral heads<br />

have decreased osteoclast number but increased <strong>bone</strong><br />

resorption when treated with IGF-I. This ambiguity <strong>between</strong><br />

the different age groups should be investigated further but<br />

may primarily result from the different developmental<br />

stages <strong>of</strong> the femoral head, suggesting an increased activity<br />

<strong>of</strong> osteoclasts in 6-week-old mice, where the secondary<br />

ossification center might initiate. However, the literature


276 Cartilage 2(3)<br />

supports the increased resorption in 6-week-old mice. One<br />

study indicates that IGF-I regulates osteoclastogenesis,<br />

which subsequently results in increased <strong>bone</strong> resorption,<br />

and that IGF-I is required for maintaining the normal interaction<br />

<strong>between</strong> osteoclasts and osteoblasts through RANKL<br />

and RANK. 43 Another study indicates that IGF-I stimulates<br />

both resorption and formation <strong>of</strong> <strong>bone</strong>. 44<br />

The concentration <strong>of</strong> CTX-I, as a function <strong>of</strong> OSM +<br />

TNF-α stimulation, increased with increasing age, which is<br />

consistent with the increasing size <strong>of</strong> the <strong>bone</strong> compartment<br />

shown by the histology. We expect the majority <strong>of</strong> CTX-I<br />

to come from <strong>bone</strong> resorption because it is mediated by<br />

cathepsin K, which is produced by the osteoclasts in this<br />

model. Correspondingly, the cartilage compartment<br />

decreased proportionally with the increasing age <strong>of</strong> the<br />

mice. However, our study showed that it was still possible<br />

to measure the anabolic and catabolic pattern <strong>of</strong> total collagen<br />

type II formation (PIINP) in 9- and 12-week-old<br />

mice, as we saw in 3-week-old mice. The formation <strong>of</strong> collagen<br />

type II decreased over time for all 3 treatments.<br />

However, IGF-I slowed the drop <strong>of</strong> the formation. This<br />

indicates that the total increase in collagen type II formation,<br />

seen by IGF-I in the bar graph (Fig. 6A), was mediated<br />

by a protective effect instead <strong>of</strong> an anabolic effect. The<br />

same protective effect <strong>of</strong> IGF-I stimulation has been published<br />

for bovine cartilage explants. 21 Another possibility is<br />

that the formation <strong>of</strong> collagen type II peaks at day 4; thus,<br />

we only detect a decrease over time. OSM + TNF-α stimulation<br />

increased the degradation <strong>of</strong> collagen type II (CTX-II)<br />

in femoral heads <strong>of</strong> all 4 age groups, which correlates with<br />

earlier ex vivo experiments stimulated with OSM + TNF-α,<br />

which showed that collagen type II degraded. 22,45 The concentration<br />

<strong>of</strong> CTX-II fragments released to the conditioned<br />

medium from the femoral heads decreased over time in<br />

3-week-old mice, probably due to a peak at day 4 since the<br />

OSM + TNF-α stimulation was very high at that particular<br />

day. Conversely, the concentrations <strong>of</strong> CTX-II in the other<br />

age groups increased over time and proportionally with the<br />

age, even though the ratio <strong>of</strong> cartilage to <strong>bone</strong> became<br />

smaller. This suggests that the peaks for CTX-II release<br />

come later in mice older than 3 weeks. Furthermore, this<br />

indicates that the secondary ossification center plays an<br />

<strong>important</strong> part in the release <strong>of</strong> CTX-II. Collagen type II is<br />

the predominant collagen <strong>of</strong> cartilage. Thus, we expect the<br />

biochemical markers, PIINP and CTX-II, to primarily<br />

come from the cartilage compartment and not from the<br />

<strong>bone</strong> or ligamentum teres.<br />

In the present study, the biochemical markers in combination<br />

with histology have shown that we now have a<br />

simple ex vivo model that henceforth can be used for<br />

screening compounds <strong>of</strong> interest. Furthermore, this model<br />

could be essential as a preliminary study to in vivo animal<br />

studies to narrow down expenses and time spent on big<br />

animal studies with futile outcomes.<br />

63<br />

In conclusion, the murine femoral head model we developed<br />

comprises both <strong>bone</strong> and cartilage in one unit and may<br />

be <strong>of</strong> particular relevance for investigating the communication<br />

<strong>between</strong> the different cell types in these 2 tissues. This<br />

might aid the understanding <strong>of</strong> diseases dealing with miscommunication<br />

or improve the efficacy for finding new<br />

therapies targeting either cartilage or <strong>bone</strong> or both. Our<br />

model enables observations not only <strong>of</strong> the cells in in situlike<br />

conditions but also monitoring <strong>of</strong> changes in <strong>bone</strong> and<br />

cartilage as mice age and in response to catabolic and anabolic<br />

stimulation. As the morphology <strong>of</strong> the femoral head<br />

changes with age, our study indicates it is critical to select<br />

the appropriate age <strong>of</strong> mice to investigate a specific question.<br />

Acknowledgments and Funding<br />

The authors gratefully acknowledge the funding from the Danish<br />

Research Foundation (Den Danske Forskningsfond) for supporting<br />

this work. The salary <strong>of</strong> the corresponding author is supported<br />

in part by The Ministry <strong>of</strong> Science Technology and Innovation <strong>of</strong><br />

Denmark.<br />

Declaration <strong>of</strong> Conflicting Interests<br />

All authors declare that the affiliation reveals full disclosure. In<br />

addition, Dr. Karsdal owns stock in Nordic Bioscience.<br />

References<br />

1. Felson DT, Neogi T. Osteoarthritis: is it a disease <strong>of</strong> cartilage<br />

or <strong>of</strong> <strong>bone</strong>? Arthritis Rheum. 2004;50(2):341-4.<br />

2. Radin EL, Rose RM. Role <strong>of</strong> <strong>subchondral</strong> <strong>bone</strong> in the initiation<br />

and progression <strong>of</strong> cartilage damage. Clin Orthop Relat<br />

Res. 1986;213:34-40.<br />

3. Dieppe P, Cushnaghan J, Young P, Kirwan J. Prediction <strong>of</strong> the<br />

progression <strong>of</strong> joint space narrowing in osteoarthritis <strong>of</strong> the knee<br />

by <strong>bone</strong> scintigraphy. Ann Rheum Dis. 1993;52(8):557-63.<br />

4. Ratcliffe A, Seibel MJ. Biochemical markers <strong>of</strong> osteoarthritis.<br />

Curr Opin Rheumatol. 1990;2(5):770-6.<br />

5. Rogers J, Shepstone L, Dieppe P. Is osteoarthritis a systemic<br />

disorder <strong>of</strong> <strong>bone</strong>? Arthritis Rheum. 2004;50(2):452-7.<br />

6. Wohl GR, Shymkiw RC, Matyas JR, Kloiber R, Zernicke RF.<br />

Periarticular cancellous <strong>bone</strong> changes following anterior cruciate<br />

ligament injury. J Appl Physiol. 2001;91(1):336-42.<br />

7. Boyd SK, Matyas JR, Wohl GR, Kantzas A, Zernicke RF.<br />

Early regional adaptation <strong>of</strong> periarticular <strong>bone</strong> mineral density<br />

after anterior cruciate ligament injury. J Appl Physiol.<br />

2000;89(6):2359-64.<br />

8. Boyd SK, Muller R, Zernicke RF. Mechanical and architectural<br />

<strong>bone</strong> adaptation in early stage experimental osteoarthritis.<br />

J Bone Miner Res. 2002;17(4):687-94.<br />

9. Burr DB, Radin EL. Micr<strong>of</strong>ractures and microcracks in <strong>subchondral</strong><br />

<strong>bone</strong>: are they relevant to osteoarthrosis? Rheum Dis<br />

Clin North Am. 2003;29(4):675-85.<br />

10. Dedrick DK, Goulet R, Huston L, Goldstein SA, Bole GG.<br />

Early <strong>bone</strong> changes in experimental osteoarthritis using microscopic<br />

computed tomography. J Rheumatol Suppl. 1991;27:44-5.


Madsen et al. 277<br />

11. Dedrick DK, Goldstein SA, Brandt KD, O’Connor BL, Goulet<br />

RW, Albrecht M. A longitudinal study <strong>of</strong> <strong>subchondral</strong><br />

plate and trabecular <strong>bone</strong> in cruciate-deficient dogs with<br />

osteoarthritis followed up for 54 months. Arthritis Rheum.<br />

1993;36(10):1460-7.<br />

12. Hayami T, Pickarski M, Wesolowski GA, McLane J, Bone<br />

A, Destefano J, et al. The role <strong>of</strong> <strong>subchondral</strong> <strong>bone</strong> remodeling<br />

in osteoarthritis: reduction <strong>of</strong> cartilage degeneration and<br />

prevention <strong>of</strong> osteophyte formation by alendronate in the rat<br />

anterior cruciate ligament transection model. Arthritis Rheum.<br />

2004;50(4):1193-206.<br />

13. Westacott CI, Webb GR, Warnock MG, Sims JV, Elson CJ.<br />

Alteration <strong>of</strong> cartilage metabolism by cells from osteoarthritic<br />

<strong>bone</strong>. Arthritis Rheum. 1997;40(7):1282-91.<br />

14. Sanchez C, Deberg MA, Piccardi N, Msika P, Reginster JY,<br />

Henrotin YE. Subchondral <strong>bone</strong> osteoblasts induce phenotypic<br />

changes in human osteoarthritic chondrocytes. Osteoarthritis<br />

Cartilage. 2005;13(11):988-97.<br />

15. Sanchez C, Deberg MA, Piccardi N, Msika P, Reginster JY,<br />

Henrotin YE. Osteoblasts from the sclerotic <strong>subchondral</strong> <strong>bone</strong><br />

downregulate aggrecan but upregulate metalloproteinases expression<br />

by chondrocytes: this effect is mimicked by interleukin-6,<br />

-1beta and oncostatin M pre-treated non-sclerotic osteoblasts.<br />

Osteoarthritis Cartilage. 2005;13(11):979-87.<br />

16. Takahashi N, Udagawa N, Takami M, Suda T. Cells <strong>of</strong> <strong>bone</strong>:<br />

osteoclast generation. In: Bilezikian JP, Raisz LG, Rodan GA,<br />

editors. Principles <strong>of</strong> <strong>bone</strong> biology. Vol. 1. 2nd ed. San Diego:<br />

Academic Press; 2002. p. 109-26.<br />

17. Fazzalari NL, Kuliwaba JS, Atkins GJ, Forwood MR, Findlay<br />

DM. The ratio <strong>of</strong> messenger RNA levels <strong>of</strong> receptor<br />

activator <strong>of</strong> nuclear factor kappaB ligand to osteoprotegerin<br />

correlates with <strong>bone</strong> remodeling indices in normal human<br />

cancellous <strong>bone</strong> but not in osteoarthritis. J Bone Miner Res.<br />

2001;16(6):1015-27.<br />

18. Bonde M, Qvist P, Fledelius C, Riis BJ, Christiansen C. Immunoassay<br />

for quantifying type I collagen degradation products<br />

in urine evaluated. Clin Chem. 1994;40(11 Pt 1):2022-5.<br />

19. Christgau S, Garnero P, Fledelius C, Moniz C, Ensig M,<br />

Gineyts E, et al. Collagen type II C-telopeptide fragments as<br />

an index <strong>of</strong> cartilage degradation. Bone. 2001;29(3):209-15.<br />

20. Schaller S, Henriksen K, Hoegh-Andersen P, Sondergaard<br />

BC, Sumer EU, Tanko LB, et al. In vitro, ex vivo, and in vivo<br />

methodological approaches for studying therapeutic targets <strong>of</strong><br />

osteoporosis and degenerative joint diseases: how biomarkers<br />

can assist? Assay Drug Dev Technol. 2005;3(5):553-80.<br />

21. Madsen SH, Sondergaard BC, Bay-Jensen AC, Karsdal MA.<br />

Cartilage formation measured by a novel PIINP assay suggests<br />

that IGF-I does not stimulate but maintains cartilage formation<br />

ex vivo. Scand J Rheumatol. 2009;38(3):222-6.<br />

22. Sondergaard BC, Henriksen K, Wulf H, Oestergaard S,<br />

Schurigt U, Brauer R, et al. Relative contribution <strong>of</strong> matrix<br />

metalloprotease and cysteine protease activities to cytokinestimulated<br />

articular cartilage degradation. Osteoarthritis Cartilage.<br />

2006;14(8):738-48.<br />

64<br />

23. Glasson SS, Askew R, Sheppard B, Carito BA, Blanchet T,<br />

Ma HL, et al. Characterization <strong>of</strong> and osteoarthritis susceptibility<br />

in ADAMTS-4-knockout mice. Arthritis Rheum.<br />

2004;50(8):2547-58.<br />

24. Hoegh-Andersen P, Tanko LB, Andersen TL, Lundberg CV,<br />

Mo JA, Heegaard AM, et al. Ovariectomized rats as a model<br />

<strong>of</strong> postmenopausal osteoarthritis: validation and application.<br />

Arthritis Res Ther. 2004;6(2):R169-80.<br />

25. Olsen AK, Sondergaard BC, Byrjalsen I, Tanko LB, Christiansen<br />

C, Muller A, et al. Anabolic and catabolic function <strong>of</strong><br />

chondrocyte ex vivo is reflected by the metabolic processing <strong>of</strong><br />

type II collagen. Osteoarthritis Cartilage. 2007;15(3):335-42.<br />

26. Bay-Jensen AC, Andersen TL, Charni-Ben TN, Kristensen<br />

PW, Kjaersgaard-Andersen P, Sandell L, et al. Biochemical<br />

markers <strong>of</strong> type II collagen breakdown and synthesis are positioned<br />

at specific sites in human osteoarthritic knee cartilage.<br />

Osteoarthritis Cartilage. 2008;16(5):615-23.<br />

27. Oettmeier R, Abendroth K. Osteoarthritis and <strong>bone</strong>: osteologic<br />

types <strong>of</strong> osteoarthritis <strong>of</strong> the hip. Skeletal Radiol.<br />

1989;18(3):165-74.<br />

28. Qvist P, Bay-Jensen AC, Christiansen C, Dam EB, Pastoureau<br />

P, Karsdal MA. The disease modifying osteoarthritis drug<br />

(DMOAD): is it in the horizon? Pharmacol Res. 2008;58(1):1-7.<br />

29. Keraplis AC. Embryonic development <strong>of</strong> <strong>bone</strong> and the molecular<br />

regulation <strong>of</strong> intramembranous and endochondral <strong>bone</strong><br />

formation. In: Bilezikian JP, Raisz LG, Rodan GA, editors.<br />

Principles <strong>of</strong> <strong>bone</strong> biology. Vol. 1. 2nd ed. San Diego: Academic<br />

Press; 2002. p. 33-58.<br />

30. von der MK, Kirsch T, Nerlich A, Kuss A, Weseloh G, Gluckert<br />

K, et al. Type X collagen synthesis in human osteoarthritic<br />

cartilage: indication <strong>of</strong> chondrocyte hypertrophy. Arthritis<br />

Rheum. 1992;35(7):806-11.<br />

31. Karsdal MA, Madsen SH, Christiansen C, Henriksen K,<br />

Fosang AJ, Sondergaard BC. Cartilage degradation is fully<br />

reversible in the presence <strong>of</strong> aggrecanase but not matrix metalloproteinase<br />

activity. Arthritis Res Ther. 2008;10(3):R63.<br />

32. Nilsson O, Marino R, De LF, Phillip M, Baron J. Endocrine<br />

regulation <strong>of</strong> the growth plate. Horm Res. 2005;64(4):157-65.<br />

33. Hutchison MR, Bassett MH, White PC. Insulin-like growth<br />

factor-I and fibroblast growth factor, but not growth hormone,<br />

affect growth plate chondrocyte proliferation. Endocrinology.<br />

2007;148(7):3122-30.<br />

34. Pass C, Macrae VE, Ahmed SF, Farquharson C. Inflammatory<br />

cytokines and the GH/IGF-I axis: novel actions on <strong>bone</strong><br />

growth. Cell Biochem Funct. 2009;27(3):119-27.<br />

35. Thomson BM, Mundy GR, Chambers TJ. Tumor necrosis factors<br />

alpha and beta induce osteoblastic cells to stimulate osteoclastic<br />

<strong>bone</strong> resorption. J Immunol. 1987;138(3):775-9.<br />

36. Kobayashi K, Takahashi N, Jimi E, Udagawa N, Takami M,<br />

Kotake S, et al. Tumor necrosis factor alpha stimulates osteoclast<br />

differentiation by a mechanism independent <strong>of</strong> the ODF/<br />

RANKL-RANK interaction. J Exp Med. 2000;191(2):275-86.<br />

37. Weir EC, Horowitz MC, Baron R, Centrella M, Kacinski<br />

BM, Insogna KL. Macrophage colony-stimulating factor


278 Cartilage 2(3)<br />

release and receptor expression in <strong>bone</strong> cells. J Bone Miner Res.<br />

1993;8(12):1507-18.<br />

38. Felix R, Fleisch H, Elford PR. Bone-resorbing cytokines enhance<br />

release <strong>of</strong> macrophage colony-stimulating activity by the osteoblastic<br />

cell MC3T3-E1. Calcif Tissue Int. 1989;44(5):356-60.<br />

39. Sato K, Kasono K, Fujii Y, Kawakami M, Tsushima T,<br />

Shizume K. Tumor necrosis factor type alpha (cachectin)<br />

stimulates mouse osteoblast-like cells (MC3T3-E1) to produce<br />

macrophage-colony stimulating activity and prostaglandin<br />

E2. Biochem Biophys Res Commun. 1987;145(1):323-9.<br />

40. Heymann D, Rousselle AV. gp130 cytokine family and <strong>bone</strong><br />

cells. Cytokine. 2000;12(10):1455-68.<br />

41. Tamura T, Udagawa N, Takahashi N, Miyaura C, Tanaka S,<br />

Yamada Y, et al. Soluble interleukin-6 receptor triggers osteoclast<br />

formation by interleukin 6. Proc Natl Acad Sci U S A.<br />

1993;90(24):11924-8.<br />

42. Richards CD, Langdon C, Deschamps P, Pennica D, Shaughnessy<br />

SG. Stimulation <strong>of</strong> osteoclast differentiation in vitro by<br />

mouse oncostatin M, leukaemia inhibitory factor, cardiotrophin-1<br />

and interleukin 6: synergy with dexamethasone. Cytokine. 2000;<br />

12(6):613-21.<br />

43. Wang Y, Nishida S, Elalieh HZ, Long RK, Halloran BP, Bikle<br />

DD. Role <strong>of</strong> IGF-I signaling in regulating osteoclastogenesis.<br />

J Bone Miner Res. 2006;21(9):1350-8.<br />

44. Chihara K, Sugimoto T. The action <strong>of</strong> GH/IGF-I/IGFBP in<br />

osteoblasts and osteoclasts. Horm Res. 1997;48(Suppl 5):<br />

45-9.<br />

45. Karsdal MA, Sumer EU, Wulf H, Madsen SH, Christiansen C,<br />

Fosang AJ, et al. Induction <strong>of</strong> increased cAMP levels in articular<br />

chondrocytes blocks matrix metalloproteinase-mediated<br />

cartilage degradation, but not aggrecanase-mediated cartilage<br />

degradation. Arthritis Rheum. 2007;56(5):1549-58.<br />

65


CHAPTER 4: PAPER I<br />

66


CHAPTER 5<br />

CHAPTER 5: PAPER II<br />

PAPER II<br />

___________________________________________________________________________<br />

Introductory remarks<br />

This manuscript shows the effect <strong>of</strong> glucocorticoids on <strong>bone</strong> and/or cartilage, performed in pre-<br />

clinical models. The novel ex vivo femoral head model (PAPER I) was used to investigate the<br />

effect <strong>of</strong> glucocorticoids on a <strong>bone</strong>/cartilage unit, whereas the articular cartilage ex vivo model and<br />

the osteoclast and osteoblast cell cultures were used to investigate the effects on either <strong>bone</strong> or<br />

cartilage. This manuscript highlights the importance <strong>of</strong> the individual cell type and verifies the<br />

novel ex vivo femoral head model as a useful screening model for potential drugs.<br />

67


Glucocorticoids exert context-dependent effects on cells <strong>of</strong> the joint in vitro<br />

Suzi H. Madsen a,⇑ , Kim V. Andreassen b , Søren T. Christensen c , Morten A. Karsdal a ,<br />

Francis M. Sverdrup d , Anne-Christine Bay-Jensen a , Kim Henriksen b<br />

a Cartilage Biology and Biomarkers, Nordic Bioscience A/S, Herlev Hovedgade 207, DK-2730 Herlev, Denmark<br />

b Bone Biology, Nordic Bioscience A/S, Herlev Hovedgade 207, DK-2730 Herlev, Denmark<br />

c Department <strong>of</strong> Biology, The August Krogh Building, University <strong>of</strong> Copenhagen, Universitetsparken 13, DK-2100 Copenhagen OE, Denmark<br />

d Center for World Health and Medicine, Saint Louis University, Doisy Research Rm 355, 1205 Carr Ln, Saint Louis, MO 63104, USA<br />

article info<br />

Article history:<br />

Received 21 June 2011<br />

Received in revised form 27 July 2011<br />

Accepted 28 July 2011<br />

Available online 10 August 2011<br />

Keywords:<br />

Prednisolone<br />

Dexamethasone<br />

Chondrocytes<br />

Osteoclasts<br />

Osteoblasts<br />

Ex vivo model<br />

1. Introduction<br />

abstract<br />

Glucocorticoids (GCs) are used to overcome inflammatory conditions,<br />

such as inflammatory bowel diseases and rheumatoid<br />

Abbreviations: IGF-I, insulin-like growth factor 1; BMP-2, <strong>bone</strong> morphogenetic<br />

protein 2; OSM, oncostatin M; TNF-a, tumor necrosis factor-a; GC, glucocorticoid;<br />

OA, osteoarthritis; RANKL, receptor activator <strong>of</strong> nuclear factor kappa-B ligand; OPG,<br />

osteoprotegerin; PRED, prednisolone; DEX, dexamethasone; MMP, matrix metalloproteinase;<br />

PBS, phosphate buffered saline; M-CSF, macrophage colony-stimulating<br />

factor; a-MEM, a-modified eagle medium; ELISA, enzyme-linked immunosorbent<br />

assay; TRAP, tartrate-resistant acid phosphatase; sGAG, sulfated glycosaminoglycans.<br />

⇑ Corresponding author. Tel.: +45 44 52 52 52; fax: +45 44 52 52 51.<br />

E-mail addresses: shm@nordicbioscience.com (S.H. Madsen), kan@nordicbioscience.com<br />

(K.V. Andreassen), stchristensen@bio.ku.dk (S.T. Christensen), mk@nordicbioscience.com<br />

(M.A. Karsdal), fsverdrup@charter.net (F.M. Sverdrup),<br />

acbj@nordicbioscience.com (A.-C. Bay-Jensen), kh@nordicbioscience.com<br />

(K. Henriksen).<br />

0039-128X/$ - see front matter Ó 2011 Elsevier Inc. All rights reserved.<br />

doi:10.1016/j.steroids.2011.07.018<br />

Steroids 76 (2011) 1474–1482<br />

Contents lists available at SciVerse ScienceDirect<br />

Steroids<br />

journal homepage: www.elsevier.com/locate/steroids<br />

Introduction: Glucocorticoids are known to attenuate <strong>bone</strong> formation in vivo leading to decreased <strong>bone</strong><br />

volume and increased risk <strong>of</strong> fractures, whereas effects on the joint tissue are less characterized. However,<br />

glucocorticoids appear to have a reducing effect on inflammation and pain in osteoarthritis. This<br />

study aimed at characterizing the effect <strong>of</strong> glucocorticoids on chondrocytes, osteoclasts, and osteoblasts.<br />

Experimental: We used four model systems to investigate how glucocorticoids affect the cells <strong>of</strong> the joint;<br />

two intact tissues (femoral head- and cartilage-explants), and two separate cell cultures <strong>of</strong> osteoblasts<br />

(2T3-pre-osteoblasts) and osteoclasts (CD14 + -monocytes). The model systems were cultured in the presence<br />

<strong>of</strong> two glucocorticoids; prednisolone or dexamethasone. To induce anabolic and catabolic conditions,<br />

cultures were activated by insulin-like growth factor I/<strong>bone</strong> morphogenetic protein 2 and<br />

oncostatin M/tumor necrosis factor-a, respectively. Histology and markers <strong>of</strong> <strong>bone</strong>- and cartilage-turnover<br />

were used to evaluate effects <strong>of</strong> glucocorticoid treatment.<br />

Results: Prednisolone treatment decreased collagen type-II degradation in immature cartilage, whereas<br />

glucocorticoids did not affect collagen type-II in mature cartilage. Glucocorticoids had an anti-catabolic<br />

effect on catabolic-activated cartilage from a bovine stifle joint and murine femoral heads. Glucocorticoids<br />

decreased viability <strong>of</strong> all <strong>bone</strong> cells, leading to a reduction in osteoclastogenesis and <strong>bone</strong> resorption;<br />

however, <strong>bone</strong> morphogenetic protein 2-stimulated osteoblasts increased <strong>bone</strong> formation, as<br />

opposed to non-stimulated osteoblasts.<br />

Conclusions: Using highly robust in vitro models <strong>of</strong> <strong>bone</strong> and cartilage turnover, we suggest that effects <strong>of</strong><br />

glucocorticoids highly depend on the activation and differential stage <strong>of</strong> the cell targeted in the joint.<br />

Present data indicated that glucocorticoid treatment may be beneficial for articular cartilage, although<br />

detrimental effects on <strong>bone</strong> should be taken into account.<br />

Ó 2011 Elsevier Inc. All rights reserved.<br />

68<br />

arthritis [1]. In addition, intra-articular injections <strong>of</strong> GCs have been<br />

tested for their ability to reduce inflammation and pain in<br />

osteoarthritis (OA) and appear to have a reducing effect on both<br />

parameters [2]. Furthermore, OA chondrocytes express fewer glucocorticoid<br />

receptors (GR) than normal chondrocytes, indicating<br />

the importance <strong>of</strong> glucocorticoid stimulation <strong>of</strong> normal cartilage<br />

[3]. However, GC treatment in children and adolescents is associated<br />

with severe growth retardation caused by apoptosis <strong>of</strong> the<br />

growth plate cartilage, especially in the proliferative zones <strong>of</strong> the<br />

cartilage [4,5]. With respect to the effects on articular chondrocytes,<br />

in vitro and ex vivo studies <strong>of</strong> chondrocytes are contradicting<br />

and highly context dependent [6–9].<br />

Unfortunately, prolonged GC therapy has also been associated<br />

with <strong>bone</strong> loss and fractures, resulting in severe osteoporosis<br />

[10,11]. In this scenario, GCs cause significant reductions in <strong>bone</strong><br />

formation, as well as a transient acceleration <strong>of</strong> <strong>bone</strong> resorption,


leading to fragility fractures [12], which especially occur in highturnover<br />

<strong>bone</strong> compartments, such as vertebrae [13]. In vivo, this<br />

effect is mediated via induction <strong>of</strong> osteoblast and osteocytes<br />

apoptosis, as well as a less well-understood activation <strong>of</strong> osteoclasts<br />

[1,14–17]. Interestingly, a mouse study showed that the<br />

detrimental effect <strong>of</strong> GCs on <strong>bone</strong> formation was mediated<br />

through the osteoclasts; whether it was dependent on <strong>bone</strong><br />

resorption was not clear [17]. In vitro studies <strong>of</strong> the effects <strong>of</strong><br />

GCs on osteoclasts have shown highly context dependent effects,<br />

i.e., both inhibitory and activating effects were observed [17–19].<br />

One possible explanation for these contradicting results may lay<br />

within the heterogeneity <strong>of</strong> the model systems used, which comprise<br />

both mouse and human cells in either purified or mixed cell<br />

populations [17–19]. In osteoblasts and their precursors, GCs are<br />

known to stimulate Receptor Activator <strong>of</strong> Nuclear Factor Kappa-B<br />

ligand (RANKL), while reducing osteoprotegerin (OPG) [18],<br />

potentially explaining the transient activation <strong>of</strong> <strong>bone</strong> resorption<br />

observed in vivo. GCs also possess the ability to stimulate in vitro<br />

osteoblastogenesis in cell cultures [20,21], although separate<br />

studies show that GCs may arrest osteoblastogenesis and introduce<br />

apoptosis [22,23], illustrating the complex and context<br />

dependent nature <strong>of</strong> GCs.<br />

Finally, GC signaling in osteoblasts and osteocytes was shown to<br />

be involved in the pathogenesis <strong>of</strong> inflammatory arthritis, demonstrating<br />

the importance <strong>of</strong> cellular cross-talk in the joint in relation<br />

to effects <strong>of</strong> GCs [24]. So, whether GCs administered in vivo affect<br />

both <strong>bone</strong> and cartilage, or just <strong>bone</strong>, is presently not clear<br />

[25,26]. Especially in OA, a disease <strong>of</strong> both <strong>bone</strong> and cartilage<br />

[27,28], the effects <strong>of</strong> GCs in the context <strong>of</strong> a whole joint need to<br />

be studied in much further detail.<br />

In this study, we used four highly robust cell systems, namely<br />

murine femoral head explants [29], bovine articular cartilage explants<br />

[30], CD14 + derived human osteoclasts [31] and the 2T3<br />

osteoblast cell line [32], to characterize the effects <strong>of</strong> the GCs –<br />

prednisolone (PRED) and dexamethasone (DEX) – on cells <strong>of</strong> the<br />

joint. Furthermore, we cultured the cells under different conditions<br />

to shed light on the context dependency <strong>of</strong> the effects <strong>of</strong><br />

GCs.<br />

2. Experimental<br />

2.1. Ex vivo models<br />

2.1.1. Murine femoral heads<br />

The experiment was performed in accordance with relevant<br />

guidelines and regulations from the Ethical Committee, as mice<br />

were euthanized before the experiment started. Femoral heads<br />

from 12-week-old outbred Naval Medical Research Institute<br />

(NMRI) female mice (Charles River, Germany) were isolated and<br />

cultured accordingly to Madsen et al. [29]. Femoral heads were cultured<br />

for 3 weeks, in the presence or absence <strong>of</strong> different factors:<br />

the anabolic factor Insulin-like Growth Factor I (IGF-I) [100 ng/<br />

ml] (Sigma, DK), the catabolic cytokines Oncostatin M (OSM)<br />

[10 ng/ml] (Sigma, DK) + Tumor Necrosis Factor-a (TNF-a)<br />

[20 ng/ml] (R&D Systems, UK) and PRED [100 nM] (Sigma, DK).<br />

2.1.2. Bovine articular cartilage<br />

Bovine knees were bought at the local slaughter house<br />

(Slangerup, DK) just after slaughtering. Articular cartilage explants<br />

were isolated and cultured accordingly to our previous<br />

studies [30,33]. The explants were cultured for 3 weeks, in the<br />

presence or absence <strong>of</strong> different factors: OSM [10 ng/ml] + TNFa<br />

[20 ng/ml], IGF-I [100 ng/ml], PRED [100 nM] and DEX<br />

[100 nM] (Sigma, DK).<br />

S.H. Madsen et al. / Steroids 76 (2011) 1474–1482 1475<br />

69<br />

2.2. In vitro models<br />

2.2.1. Human osteoclast preparation<br />

Human blood was donated from the Danish University Hospital<br />

<strong>of</strong> Copenhagen (has an approval from the Ethical Committee to donate<br />

blood to us), and CD14 + monocytes were isolated and differentiated<br />

into osteoclasts accordingly to Sorensen et al. [31].<br />

2.2.2. Differentiating osteoclasts<br />

The cells were seeded on plastic or bovine cortical <strong>bone</strong> slices<br />

(Nordic Bioscience Diagnostics, DK) at a density <strong>of</strong> 100,000 cells/<br />

well in a 96-well plate and cultured in phenol red free a-MEM containing<br />

10% serum, P/S, M-CSF [25 ng/ml] (R&D System, DK),<br />

RANKL [25 ng/ml] (R&D Systems, DK), either in the presence or absence<br />

<strong>of</strong> DEX [100 nM] (Sigma, DK). Incubation was performed at<br />

37 °C and 5% CO2. The media were changed every 2nd or 3rd day<br />

and saved at 20 °C.<br />

2.2.3. Mature osteoclasts<br />

For experiments with mature human osteoclasts, cells were<br />

grown in 75 cm 2 culture flasks for 10 days in phenol red free a-<br />

MEM containing 10% serum, P/S, M-CSF [25 ng/ml], and RANKL<br />

[25 ng/ml] to induce osteoclastogenesis until the cells had 3–5 nuclei<br />

on average. The cells were washed with PBS twice, trypsin was<br />

added, and the cells were incubated at 37 °C for approximately<br />

20 min. The cells were scraped <strong>of</strong>f, and re-seeded on plastic or bovine<br />

cortical <strong>bone</strong> slices at a density <strong>of</strong> 40,000 cells/well in a 96well<br />

plate in the presence <strong>of</strong> phenol red free a-MEM containing<br />

10% serum, P/S, M-CSF [25 ng/ml], RANKL [25 ng/ml] in the presence<br />

or absence <strong>of</strong> DEX [100 nM] at 37 °C and 5% CO2. The media<br />

were changed every 2nd or 3rd day and saved at 20 °C.<br />

2.2.4. Murine osteoblasts<br />

2T3 pre-osteoblast cells were purchased from Dr. Steven E. Harris<br />

at the University <strong>of</strong> Texas Health Science Center at San Antonio.<br />

The murine mesenchymal stem cell line, 2T3, is able to differentiate<br />

into osteoblasts under proper predetermined conditions. Osteoblasts<br />

were prepared accordingly to Ghosh-Choudhury et al. [32].<br />

To induce <strong>bone</strong> nodule formation, the 2T3 cells were lifted by trypsin<br />

and reseeded in 24-well plates at a density <strong>of</strong> 10,000 cells/well,<br />

and then cultured in the presence <strong>of</strong> ascorbic acid [100 lg/ml] and<br />

b-glycerol-phosphate [4 mM], in the presence or absence <strong>of</strong> BMP-2<br />

[30 ng/ml] or DEX [100 nM] for 10 days. Incubation was performed<br />

at 37 °C, 5% CO2. The media were replaced every 2nd or 3rd day<br />

and saved at 20 °C.<br />

2.3. Concentrations<br />

The concentrations <strong>of</strong> IGF-I, BMP-2 and OSM + TNF-a were selected<br />

accordingly to previous studies [29–31,33,34]. The concentrations<br />

<strong>of</strong> DEX and PRED were selected accordingly to previous<br />

studies [5,6,17].<br />

2.4. Cell-survival after culture-period<br />

The viability <strong>of</strong> the cells (in explants or isolated cells) were measured<br />

at the end <strong>of</strong> the culture period, using the colorimetric dye,<br />

alamar blue (Invitrogen, DK), as a 10% solution in the conditioned<br />

media, accordingly to the manufacturer’s instructions [35].<br />

2.5. Biomarkers<br />

The resorption marker CTX-I (RatLaps Ò ), the <strong>bone</strong> formation<br />

marker Rat/Mouse PINP EIA and the cartilage formation marker<br />

PIINP (IDS Ltd., UK) were measured accordingly to the manufacturer’s<br />

instructions [33]. The competitive ELISA CIIM (Nordic


1476 S.H. Madsen et al. / Steroids 76 (2011) 1474–1482<br />

Bioscience, DK) was used to quantify MMP-mediated degradation<br />

products <strong>of</strong> type II collagen, accordingly to Bay-Jensen et al. [36].<br />

342-G2 measures MMP-mediated aggrecan degradation accordingly<br />

to Sumer et al. [37].<br />

2.6. Quantification <strong>of</strong> osteoclast number<br />

The TRAP assay was performed as previously described [31].<br />

2.7. Measurements <strong>of</strong> alkaline phosphatase activity<br />

The culture supernatants were added to a substrate mixture <strong>of</strong><br />

95% AMP buffer (pH 10), 10.1 mM phosphatase substrate, 18 mM<br />

MgCl2, and MilliQ in a ratio <strong>of</strong> 1:4 and incubated in the dark for<br />

20 min. After incubation, the reaction was stopped suing 0.5 M<br />

NaOH, and finally the absorbance at 405 nm was measured using<br />

an ELISA reader.<br />

2.8. Resorption assays<br />

The concentration <strong>of</strong> total calcium was measured in culture<br />

supernatants after resorption, by using a colorimetric assay and a<br />

Hitachi 912 Automatic Analyzer (Roche Diagnostics) following<br />

the assay method validated and warranted by Roche Diagnostics<br />

[38].<br />

2.9. Alizarin Red-S staining <strong>of</strong> mineralized <strong>bone</strong> nodules<br />

and extraction <strong>of</strong> dye<br />

After 10 days <strong>of</strong> culture, the osteoblast cultures were stopped<br />

by fixation in 4% formaldehyde for 20 min. The cells were stained<br />

in 40 mM Alizarin Red-S, pH 4.2, for 10 min, and washed thoroughly.<br />

The dye was extracted from the cells by incubation in<br />

10% cetylpyridinium chloride for 15 min while shaking in the dark,<br />

and finally the absorbance at 561 nm was measured using an ELISA<br />

reader.<br />

2.10. Histologic analysis<br />

The bovine articular cartilage explants were fixed in 4% formaldehyde,<br />

processed, paraffin embedded, and subsequently sectioned<br />

(5 lm). Proteoglycans were stained after a standard toluidine blue<br />

protocol [39] after deparaffinization and rehydration.<br />

2.10.1. Image analysis<br />

Digital images <strong>of</strong> stained sections were displayed on a computer<br />

monitor and captured using an Olympus BX-60 microscope<br />

equipped with a 60 objective and digital camera.<br />

2.11. Statistic analysis<br />

Results are shown as mean + standard error <strong>of</strong> mean (SEM). Statistic<br />

analysis was performed by the Student’s two-tailed unpaired<br />

t-test assuming normal distribution and equal variance. Statistic<br />

significance is indicated by the number <strong>of</strong> asterisks, ⁄ p < 0.05,<br />

⁄⁄ p < 0.01, and ⁄⁄⁄ p < 0.001.<br />

3. Results<br />

3.1. Prednisolone decreases cartilage degradation and <strong>bone</strong><br />

turnover in femoral heads ex vivo<br />

Prednisolone (PRED) treatment for 2 weeks decreased overall<br />

cell viability in femoral heads by 40% (p < 0.01) compared to<br />

the vehicle control (Fig. 1A). Cell viability was not affected by<br />

70<br />

OSM + TNF-a stimulation (catabolic control) (Fig. 1A). PRED in<br />

combination with OSM + TNF-a showed a tendency towards decreased<br />

viability compared to the catabolic control (Fig. 1A).<br />

After 1 week, PRED caused a reduction in the number <strong>of</strong> osteoclasts<br />

in both resting and catabolic activated femoral heads, compared<br />

to the respective vehicle and catabolic controls (Fig. 1B).<br />

After 2 weeks in culture, osteoclast number was decreased to a<br />

low level in the vehicle control and no differences were detected<br />

with any <strong>of</strong> the treatment (Fig. 1B). After 1 week, PRED treatment<br />

decreased <strong>bone</strong> resorption by 75% (p < 0.01) (Fig. 1C), consistent<br />

with the reduced number <strong>of</strong> osteoclasts (Fig. 1B). Bone resorption<br />

increased by 5.5-fold (p < 0.01) after 1 week when stimulated<br />

with OSM + TNF-a, whereas this increase in <strong>bone</strong> resorption decreased<br />

by 90% (p < 0.05) in the presence <strong>of</strong> PRED (Fig. 1C). The<br />

increase in <strong>bone</strong> resorption mediated by OSM + TNF-a after 1<br />

week, was not seen after 2 weeks (Fig. 1C). PRED treatment decreased<br />

<strong>bone</strong> formation by 50% (p < 0.01) after 1 week, and<br />

90% (p < 0.01) after 2 weeks, compared to the respective vehicles<br />

(Fig. 1D). Furthermore, OSM + TNF-a decreased <strong>bone</strong> formation by<br />

75% (p < 0.01) after 1 week, compared to the vehicle (Fig. 1D).<br />

Treatment with PRED in combination with OSM + TNF-a did not<br />

significantly alter <strong>bone</strong> formation compared to the catabolic<br />

control; however, the combination seemed to have a tendency to<br />

decrease <strong>bone</strong> formation slightly (Fig. 1D). After 2 weeks,<br />

OSM + TNF-a stimulation inhibited <strong>bone</strong> formation, compared to<br />

the vehicle (Fig. 1D). With respect to cartilage metabolism, PRED<br />

treatment showed at tendency to decrease cartilage degradation<br />

in non-stimulated femoral heads after 1 week (by 75%) and significantly<br />

by 60% (p < 0.05) after 2 weeks (Fig. 1E). OSM + TNF-a<br />

stimulation increased the cartilage degradation by 33-fold<br />

(p < 0.01) after 1 week and 30-fold (p < 0.01) after 2 weeks, compared<br />

to respective vehicles (Fig. 1E). These catabolic-mediated increases<br />

in cartilage degradation were both inhibited by the<br />

presence <strong>of</strong> PRED ( 95% (p < 0.05)) after 1 and 2 weeks (Fig. 1E).<br />

The formation <strong>of</strong> cartilage did not significantly vary <strong>between</strong> vehicle<br />

and stimulated explants after 1 week (Fig. 1F). However,<br />

OSM + TNF-a stimulation inhibited cartilage formation by 95%<br />

(p < 0.05) after 2 weeks <strong>of</strong> culture (Fig. 1F). While PRED itself did<br />

not affect cartilage formation at one or 2 weeks, it failed to prevent<br />

the inhibition caused by 2 weeks <strong>of</strong> catabolic stimulation (Fig. 1F).<br />

3.2. Prednisolone does not decrease the formation <strong>of</strong> <strong>bone</strong> and<br />

cartilage in anabolic activated femoral heads<br />

Stimulation with IGF-I increased viability <strong>of</strong> the femoral heads<br />

after 2 weeks <strong>of</strong> culture by 1.6-fold (p < 0.01) (Fig. 2A). PRED in<br />

combination with IGF-I did not affect the viability compared to<br />

the anabolic control (IGF-I alone) (Fig. 2A). Due to the low level<br />

<strong>of</strong> TRAP, we were not able to detect any differences in the number<br />

<strong>of</strong> osteoclasts by IGF-I or IGF-I + PRED treatment compared to<br />

respective vehicles (Fig. 2B). However, IGF-I stimulation decreased<br />

<strong>bone</strong> resorption by 70% (p < 0.05) after 1 week, which was further<br />

decreased by addition <strong>of</strong> PRED (Fig. 2C). PRED also decreased the<br />

IGF-I-reduced resorption by 75% (p < 0.05) after 2 weeks<br />

(Fig. 2C). The anabolic-stimulated femoral heads showed increased<br />

<strong>bone</strong> formation by 2.1-fold (p < 0.01) after 1 week and 13.4-fold<br />

(p < 0.01) after 2 weeks (Fig. 2D). PRED did not significantly decrease<br />

anabolic activated <strong>bone</strong> formation, although a downward<br />

trend was observed after 2 weeks (Fig. 2D). IGF-I-stimulated femoral<br />

heads showed very low levels <strong>of</strong> cartilage degradation after 1<br />

and 2 weeks, due to the overall low level <strong>of</strong> degradation in this setup.<br />

However, after 1 week, IGF-I stimulation decreased by 72%<br />

(p < 0.05) compared to vehicle. Addition <strong>of</strong> PRED to the anabolic<br />

control showed the same low levels <strong>of</strong> cartilage degradation compared<br />

to the anabolic controls (Fig. 2E). Cartilage formation, on the<br />

other hand, showed a tendency to increase with IGF-I stimulation


after 1 week (Fig. 2F). The addition <strong>of</strong> PRED to IGF-I stimulation<br />

was however not different from the vehicle. On the contrary, after<br />

2 weeks, PRED in combination with IGF-I showed a tendency to increase<br />

cartilage formation compared to the anabolic control<br />

(Fig. 2F).<br />

3.3. Cytokine-induced cartilage turnover is decreased<br />

by glucocorticoid treatment<br />

The effect <strong>of</strong> PRED was tested on bovine cartilage explants.<br />

However, since the effects <strong>of</strong> PRED were weak, we also tested the<br />

more potent dexamethasone (DEX) [8] in a separate experiment.<br />

The effects <strong>of</strong> PRED and DEX were measured at day 21.<br />

After 21 days <strong>of</strong> culture, the viability <strong>of</strong> the bovine cartilage explants<br />

increased with OSM + TNF-a stimulation by 1.6-fold<br />

(p < 0.05) (Fig. 3A) and 1.7-fold (p < 0.01) (Fig. 3E), compared to<br />

respective vehicles (Fig. 3A and E). However, this increase seemed<br />

inhibited by DEX (p < 0.05) (Fig. 3E). Aggrecan degradation was induced<br />

by OSM + TNF-a by more than 150-fold (p < 0.001) for both<br />

experiments, and reduced by addition <strong>of</strong> DEX by 70% (p < 0.01);<br />

however not to the level <strong>of</strong> the vehicle or DEX alone (Fig. 3B and<br />

F). The same pattern was observed for collagen type II degradation,<br />

where OSM + TNF-a stimulation increased the degradation by<br />

more than 40-fold (p < 0.01), which was reduced by addition <strong>of</strong><br />

PRED and significantly by addition <strong>of</strong> DEX by 75% (p < 0.01)<br />

(Fig. 3C and G). Aggrecan and collagen degradation were also measured<br />

at days 5 and 12, but did not show any differences <strong>between</strong><br />

the vehicle and activated explants (data not shown). OSM + TNF-a<br />

stimulation increased cartilage formation by more than 4-fold<br />

(p < 0.01) in both experiments (Fig. 3D and H). Cartilage formation<br />

S.H. Madsen et al. / Steroids 76 (2011) 1474–1482 1477<br />

Fig. 1. The effects <strong>of</strong> prednisolone in resting and catabolic-induced <strong>bone</strong> and cartilage from femoral heads. Femoral heads were cultured in the presence <strong>of</strong> 100nM<br />

prednisolone (PRED) and/or cytokines (O + T) [10 ng/ml] + [20 ng/ml] for 2 weeks. Cell viability was measured with alamar blue after 2 weeks (A). Biomarkers were measured<br />

after 1 week (black bars) and 2 weeks (gray bars) (B–F). The number <strong>of</strong> osteoclasts was measured with TRAP activity (B). The <strong>bone</strong> resorption and formation were measured<br />

by the collagen type I markers, CTX-I (C) and PINP (D), respectively. Cartilage degradation and formation were measured by the collagen type II markers, CIIM (E) and PIINP<br />

(F), respectively. PRED [100 nM] treatment decreased viability <strong>of</strong> the cells in the femoral heads, osteoclast number, <strong>bone</strong> resorption, <strong>bone</strong> formation and cartilage degradation.<br />

Each treatment n P 4.<br />

71<br />

was also induced by IGF-I stimulation by more than 8.5-fold<br />

(p < 0.01) in both experiments (Fig. 3D and H). DEX inhibited formation<br />

induced by OSM + TNF-a by 50% (p < 0.05) (Fig. 3H). PRED<br />

or DEX by themselves could not induce cartilage formation<br />

although there was a tendency towards increased formation when<br />

combined with IGF-I (Fig. 3D and H). Generally, we saw the same<br />

pattern for both GCs. However, PRED was less potent than DEX<br />

as observed in the literature [8].<br />

The biomarker data for aggrecan degradation were partly supported<br />

by histology <strong>of</strong> the explants, using toluidine blue to detect<br />

sulfated glycosaminoglycans (sGAGs) (Fig. 3I–T). The controls<br />

worked; as shown in previous studies [30], stimulation with<br />

OSM + TNF-a depleted the cartilage explants <strong>of</strong> sGAGs (Fig. 3J and<br />

P), whereas IGF-I stimulation increased the content <strong>of</strong> sGAGs<br />

(Fig. 3K and Q), compared to respective vehicles (Fig. 3I and O).<br />

The cartilage explants treated with either PRED or DEX seemed to<br />

have less sGAGs retained in the matrixes (Fig. 3L and R) than the<br />

vehicle explants (Fig. 3I and O). On the other hand, when adding<br />

DEX to OSM + TNF-a-stimulated cartilage explant, a small portion<br />

<strong>of</strong> the sGAGs seemed to be retain in the matrix (Fig. 3S) compared<br />

to the OSM + TNF-a-stimulated explant (Fig. 3P), although the visible<br />

difference is almost lost on the digital images. Furthermore,<br />

addition <strong>of</strong> PRED or DEX to the IGF-I-stimulated cartilage explants<br />

seemed to increase the content <strong>of</strong> sGAGs (Fig. 3N and T).<br />

3.4. The effect <strong>of</strong> dexamethasone depends on the activation<br />

stage <strong>of</strong> osteoblasts<br />

To investigate the effects <strong>of</strong> GCs on <strong>bone</strong> cells, the more potent<br />

DEX [8] was used for further experiments/studies (Fig. 3). When


1478 S.H. Madsen et al. / Steroids 76 (2011) 1474–1482<br />

Fig. 2. The effects <strong>of</strong> prednisolone in anabolic-induced <strong>bone</strong> and cartilage from femoral heads. Femoral heads were cultured in the presence <strong>of</strong> 100 nM prednisolone (PRED)<br />

and/or 100 ng/ml IGF-I for 2 weeks. Cell viability was measured with alamar blue after 2 weeks (A). Biomarkers were measured after 1 week (black bars) and 2 weeks (gray<br />

bars) (B–F). The number <strong>of</strong> osteoclasts was measured with TRAP activity (B). The <strong>bone</strong> resorption and formation were measured by the collagen type I markers, CTX-I (C) and<br />

PINP (D), respectively. Cartilage degradation and formation were measured by the collagen type II markers, CIIM (E) and PIINP (F), respectively. PRED [100 nM] + IGF-I<br />

[100 ng/ml] treatment decreased <strong>bone</strong> resorption, but not <strong>bone</strong> formation. Furthermore, PRED [100 nM] + IGF-I [100 ng/ml] treatment increased cartilage formation after 2<br />

weeks. Each treatment n P 4.<br />

osteoblasts were cultured in the presence <strong>of</strong> DEX, cell viability was<br />

reduced by 55% (p < 0.05), compared to the vehicle (Fig. 4A). The<br />

same pattern was seen in the presence <strong>of</strong> both DEX and BMP-2,<br />

where viability decreased by 35% (p < 0.05), compared to BMP-<br />

2 stimulation alone (Fig. 4A). DEX also decreased the nodule formation<br />

by 65% (p < 0.05), compared to vehicle (Fig. 4B). Nodule<br />

formation was induced by addition <strong>of</strong> the positive control [34],<br />

BMP-2, by 2.4-fold (p < 0.05) compared to vehicle (Fig. 4B). Furthermore,<br />

addition <strong>of</strong> DEX to BMP-2-stimulated osteoblasts, led<br />

to a further increase in nodule formation by 1.8-fold (p < 0.05)<br />

(Fig. 4B). ALP production by the osteoblasts was reduced when cultured<br />

with DEX, both in the presence (by 45%) or absence <strong>of</strong> BMP-<br />

2 (by 85%) (p < 0.05) (Fig. 4C), correlating with the cell viability<br />

measurements (Fig. 4A).<br />

3.5. Dexamethasone decreases viability and function <strong>of</strong> both<br />

mature osteoclasts and their precursors<br />

When pre-osteoclasts were cultured either on <strong>bone</strong> or plastic,<br />

DEX reduced the viability <strong>of</strong> the cells by more than 95%<br />

(p < 0.01), as well as the osteoclast marker TRAP by more than<br />

98% (p < 0.01) (Fig. 5A and B). Furthermore, the release <strong>of</strong> calcium<br />

during resorption <strong>of</strong> <strong>bone</strong>, decreased by 85% (p < 0.01) when treated<br />

with DEX (Fig. 5C). To investigate whether this effect was specific<br />

to RANKL induction <strong>of</strong> osteoclastogenesis, an experiment<br />

omitting RANKL was conducted [31]. In these cells, we again observed<br />

a reduction in cell viability in the presence <strong>of</strong> DEX (data<br />

not shown), thus showing that the reduction in viability is related<br />

to the macrophages rather than specifically to the osteoclasts. In<br />

mature osteoclasts, DEX decreased cell viability by 80%<br />

72<br />

(p < 0.01) on both <strong>bone</strong> and plastic (Fig. 5D). The number <strong>of</strong> osteoclasts<br />

was decreased by 70% (p < 0.01) on <strong>bone</strong> and 75%<br />

(p < 0.01) on plastic (Fig. 5E). Bone resorption decreased by 40%<br />

(p < 0.05) (Fig. 5F). Interestingly, the overall level <strong>of</strong> suppression induced<br />

by the DEX on mature osteoclasts was lower than the level<br />

<strong>of</strong> suppression induced in the pre-osteoclasts.<br />

4. Discussion<br />

Intra-articular corticosteroid injections have been tested for<br />

beneficial effects on inflammation and joint destruction, and appear<br />

to have some applicability by reducing pain in OA patients<br />

[2,40,41]. However, how GCs affect the individual cell types in<br />

the joint is not known. Thus, this study focused on elucidating<br />

whether GCs have any effects on cartilage turnover. Furthermore,<br />

since <strong>subchondral</strong> <strong>bone</strong> has been shown to play an essential role<br />

in the development <strong>of</strong> OA, and since the effects <strong>of</strong> GCs on <strong>bone</strong> cells<br />

are still discussed [27,42], we extended the study by performing a<br />

thorough characterization <strong>of</strong> the effect <strong>of</strong> GCs on the two major<br />

<strong>bone</strong> cell types, namely osteoblasts and osteoclasts. This study<br />

used four model systems to investigate how GCs affect the cells<br />

<strong>of</strong> the joint both in the setting <strong>of</strong> intact tissues, as femoral heads<br />

or cartilage explants (resting, catabolically active or anabolically<br />

active), and as separate cell cultures <strong>of</strong> osteoblasts (resting or differentiating)<br />

and osteoclasts (differentiating or mature and on<br />

plastic or <strong>bone</strong>).<br />

Several studies <strong>of</strong> GC-mediated effects on chondrocyte function<br />

have been published; however no clear-cut conclusion has been<br />

presented. These studies have indicated both beneficial and detrimental<br />

effects <strong>of</strong> GCs on cartilage, [6,7] however, the studies also


use a range <strong>of</strong> assays ranging from isolated chondrocytes through<br />

ex vivo explants to mesenchymal stem cell differentiation and alginate<br />

beads. In the present study, we used a murine ex vivo femoral<br />

head model [29], and found that PRED decreased <strong>bone</strong> turnover<br />

S.H. Madsen et al. / Steroids 76 (2011) 1474–1482 1479<br />

Fig. 3. The effects <strong>of</strong> prednisolone and dexamethasone in resting and activated cartilage. Bovine articular cartilage explants were cultured in the presence <strong>of</strong> 100 nM<br />

prednisolone (PRED), 100 nM dexamethasone (DEX), and/or cytokines (O + T) [10 ng/ml] + [20 ng/ml] (gray bars) and/or IGF-I [100 ng/ml] (light gray bars) for 3 weeks. Cell<br />

viability was measured with alamar blue after 3 weeks (A and E) and biomarkers were measured at day 19 (B–D and F–H). Explants cultured for 3 weeks were stained with<br />

toluidine blue to evaluate proteoglycan content (I–N and O–T). MMP-mediated aggrecan degradation was measured with 342-G2 (B and F). Collagen type II degradation and<br />

formation were measured by the markers, CIIM (E) and PIINP (F), respectively. PRED/DEX [100 nM] treatment did not affect the cartilage. However, cytokine-induced<br />

degradation were attenuated by 100 nM PRED/DEX. PRED [100 nM] was less potent than DEX [100 nM]. Each treatment n P 5.<br />

Fig. 4. The effect <strong>of</strong> dexamethasone in non-stimulated and anabolic activated osteoblasts. Osteoblasts were cultured in the presence or absence <strong>of</strong> 100 nM dexamethasone<br />

(DEX), in non-stimulated (black bars) or 30 ng/ml BMP-2 stimulated (gray bars) cells for 10 days. Cell viability was measured with alamar blue after 10 days (A) and <strong>bone</strong><br />

formation was measured by extraction <strong>of</strong> Alizarin Red-S from stained mineralized <strong>bone</strong> nodules at day 10 (B). Osteoblastogenesis was measured by alkaline phosphatase<br />

activity in the supernatants accumulated over 10 days (C). DEX [100 nM] treatment decreased viability, <strong>bone</strong> formation and osteoblastogenesis. However, DEX [100 nM]<br />

increased <strong>bone</strong> formation in BMP-2 [30 ng/ml] stimulated osteoblasts (B). Each treatment n P 4.<br />

73<br />

while mediating anti-catabolic effects on cartilage. Whether osteoclasts,<br />

osteoblasts and chondrocytes were affected directly or indirectly<br />

by PRED, were investigated by separate cell or explants<br />

cultures. To evaluate the effect <strong>of</strong> GCs directly on chondrocytes,


1480 S.H. Madsen et al. / Steroids 76 (2011) 1474–1482<br />

Fig. 5. The effect <strong>of</strong> dexamethasone in pre-osteoclasts and mature osteoclasts. Osteoclasts were cultured in on either <strong>bone</strong> (black bars) or plastic (gray bars) in the presence or<br />

absence <strong>of</strong> 100 nM dexamethasone (DEX). Pre-osteoclasts were cultured for 21 days (A–C) and mature osteoclasts for 14 days (D–F). Cell viability was measured with alamar<br />

blue after 14 or 21 days (A and D). The number <strong>of</strong> osteoclasts was measured with TRAP activity (B and E) and <strong>bone</strong> resorption was measured by calcium release from <strong>bone</strong> (C<br />

and F). DEX [100 nM] treatment decreased cell viability, osteoclast number and <strong>bone</strong> resorption in both pre-osteoclasts and mature osteoclasts. However, the effects <strong>of</strong> DEX<br />

[100 nM] on pre-osteoclasts were higher than the effects on mature osteoclasts. Each treatment n P 5.<br />

we used the bovine articular cartilage explants model that is presently<br />

the preferred model for studying cartilage turnover ex vivo<br />

[30,33]. The strong indications that OSM + TNF-a reduced the<br />

ability to induce cartilage degradation in the presence <strong>of</strong> DEX or<br />

PRED, evaluated by biomarkers and histology, suggest a chondroprotective<br />

effect by GCs independent <strong>of</strong> <strong>bone</strong> cells. Furthermore,<br />

histology showed that GC treatment had a positive effect on proteoglycans<br />

under anabolic conditions, which is in line with a study<br />

by Van der Kraan et al. that showed that PRED stimulates proteoglycan<br />

synthesis in patellar cartilage, validated by [ 35 S] incorporation<br />

[43]. However, the catabolic stimulated cartilage explants in<br />

the presence or absence <strong>of</strong> DEX, were both close to be depleted<br />

for proteoglycans, measured by the intensity <strong>of</strong> color (sGAG),<br />

which was not as convincing as the 70% reduction <strong>of</strong> MMP-mediated<br />

aggrecan degradation shown with the biomarker, 342-G2.<br />

However, depletion <strong>of</strong> color may results from an early loss <strong>of</strong> aggrecanase-mediated<br />

highly-sGAG-saturated aggrecan degradation,<br />

leaving low-sGAG-saturated aggrecan to be degraded by MMPs at<br />

day 19 [44], as we did not see any MMP-mediated degradation at<br />

days 5 and 12. This suggests that the anti-catabolic effects <strong>of</strong> GC<br />

on cartilage may be limited to the degradation mediated by MMPs.<br />

Interestingly, GC treatment in resting conditions resulted in a small<br />

loss <strong>of</strong> proteoglycans from the articular cartilage explants that we<br />

were unable to detect using the MMP-mediated aggrecan degradation<br />

marker, 342-G2. Thus, the two opposite effects <strong>of</strong> GCs treatment<br />

(protecting in activated cartilage and degrading in resting<br />

cartilage) indicate that the activation stage <strong>of</strong> the chondrocytes is<br />

<strong>important</strong> in relation to the GC response. If the GC-mediated protective<br />

effect results from an alteration <strong>of</strong> the chondrocyte phenotype<br />

(preventing the induction/release <strong>of</strong> proteases) or that the<br />

74<br />

GCs inhibit the cytokine pathway, still remains unclear. Yet, our<br />

data indicate that a reduction <strong>of</strong> MMP activity is involved, as the<br />

MMP-dependent biomarkers, 342-G2 and CIIM, are reduced. This<br />

is in line with a recent study by Garvican et al. suggesting that<br />

GCs reduce MMP-mediated cartilage degradation [9], although<br />

other enzymatic pathways, such as the u-PA/plasminogen/plasmin-pathway,<br />

also have been indicated [45]. Thus, in the search<br />

for new disease modifying osteoarthritic drugs (DMOADs), we suggest<br />

that the inhibition <strong>of</strong> protease pathways mediated by GCs<br />

could be <strong>of</strong> interest.<br />

We did not see any protective effects <strong>of</strong> GCs on the non-stimulated<br />

bovine cartilage explants, as opposed to the results with cartilage<br />

from the femoral head explants. The bovine cartilage<br />

explants represent ‘‘healthy’’ cartilage, whereas the femoral heads<br />

consist <strong>of</strong> cartilage in development, which contains more proliferating,<br />

pre-hypertrophic and hypertrophic chondrocytes (which are<br />

observed in OA cartilage) than the bovine cartilage explants [29].<br />

This could explain why we have to induce a catabolic milieu with<br />

cytokines in the bovine cartilage explants, before we see a protective<br />

effect <strong>of</strong> GCs, and suggest that GCs mainly affect chondrocytes<br />

which are activated, whereas resting chondrocytes appear to respond<br />

less. We do acknowledge that we deal with cross-species<br />

comparisons, which may cause the different effects observed in<br />

this study. However, the anabolic and catabolic controls have the<br />

same effects on the different species, suggesting the effects <strong>of</strong><br />

GC, or lack <strong>of</strong> same, are due to action from the GC and not a species-dependent<br />

effect.<br />

The present study clearly shows that GCs have demonstrably<br />

large effects on cells <strong>of</strong> <strong>bone</strong>. While it for long has been known that<br />

GCs induce apoptosis <strong>of</strong> osteoblasts and osteocytes in vivo, there is


still some controversy regarding the mechanism <strong>of</strong> action. Some<br />

data indicate a role for osteoclasts and others data indicate direct<br />

effects on osteoblasts [17,46]. Controversially, a recent study<br />

showed that the endogenous GC-induced signaling in osteoblasts<br />

is needed for proper <strong>bone</strong> formation in vivo in mice [47]. In vitro<br />

data regarding <strong>bone</strong> formation are also confusing, as data showing<br />

that GCs support stimulation <strong>of</strong> RANKL production, increase nodule<br />

formation, as well as inhibiting <strong>bone</strong> formation [18,20,21,48]. Our<br />

data shows that GC-mediated effects on osteoblasts in cell cultures<br />

are dependent on the ‘‘activation state’’ <strong>of</strong> the osteoblasts, with a<br />

toxic effect on non-activated cells, and a pro-osteoblastic effect<br />

on BMP-2-induced cells. This is in line with the results from the<br />

femoral heads, which decreased <strong>bone</strong> formation with PRED treatment,<br />

but increased <strong>bone</strong> formation in combination with IGF-I.<br />

The conflicting in vitro and in vivo data could indicate that we need<br />

to look more closely into GCs-mediated effects in the different<br />

functional subgroups <strong>of</strong> osteoblasts, and more <strong>important</strong>ly, how<br />

to modulate these with respect to preventing GC-induced <strong>bone</strong><br />

damage [48,49].<br />

Several studies have indicated that GCs stimulate osteoclastogenesis<br />

in vitro [50,51]; however, most <strong>of</strong> these studies were conducted<br />

in the presence <strong>of</strong> stromal/osteoblastic cells, which are<br />

known to produce RANKL in response to GCs [14,18]. We found<br />

that in pure osteoclast precursor cell cultures, GCs inhibited osteoclastogenesis<br />

by reducing the viability <strong>of</strong> the cells, which is in line<br />

with the decreased viability and osteoclast number found in PREDstimulated<br />

femoral heads. These data correlate to data showing decreased<br />

osteoclastogenesis in vivo in mice, and strongly indicate<br />

that the primary effect <strong>of</strong> GCs on osteoclastogenesis is inhibitory<br />

[15,16]. However, an interesting observation in this regard is that<br />

the effect <strong>of</strong> GCs on the mature osteoclasts, on cell viability, osteoclast<br />

number and resorption, was smaller than the effect on the differentiating<br />

osteoclasts. This could indicate that osteoclasts and<br />

their precursors have different levels <strong>of</strong> sensitivity to GC treatment.<br />

As all osteoclast cultures are a mix <strong>of</strong> mature multinucleated<br />

osteoclasts and their precursor cells, and the ratio <strong>of</strong> mature to<br />

pre-osteoclastic cells is dependent on several different aspects,<br />

such as density, donor, serum, etc. [31] we speculate that the reason<br />

for the predominantly toxic effect could be that we have more<br />

osteoclast precursors present in the cultures than Soe and Delaisse,<br />

who described a prolongation <strong>of</strong> the resorption cycle, when treating<br />

mature osteoclasts derived from CD14 + cells with PRED [52].<br />

A limitation to the study is that the effects observed with DEX<br />

and PRED have not been tested for steroids specificity with a<br />

non-GC control (e.g. estrogen or cholesterol). However, Kim et al.<br />

[17] showed that GR°C / mice were spared the impact <strong>of</strong> DEX<br />

on osteoclasts and their precursors, indicating a true GR-mediated<br />

action <strong>of</strong> DEX. This steroid specificity is furthermore supported by<br />

Battista et al. [3], which showed that the effect from DEX was<br />

dependent on the percentage <strong>of</strong> the GR in the chondrocytes. Nonetheless,<br />

future studies should include a non-GC control to ensure<br />

that the effects observed are indeed specific to the GC class <strong>of</strong><br />

steroids.<br />

In conclusions, we found that GCs did not appear to affect resting<br />

chondrocyte lifespan and function. Furthermore, GCs showed a<br />

protective effect on catabolic-induced cartilage and showed a tendency<br />

to increase cartilage formation under anabolic conditions.<br />

These results are encouraging for the use <strong>of</strong> intra-articular GC<br />

injections in OA knees. However, considering the well-established<br />

consequences on <strong>bone</strong> it is <strong>important</strong> to remember the tight functional<br />

relationship <strong>between</strong> <strong>bone</strong> and cartilage under both physiologically<br />

and pathologically conditions [27]. Thus, further studies<br />

into the functional relationship <strong>between</strong> these compartments are<br />

needed, especially with the focus on the possibility to rescue <strong>bone</strong><br />

damage by the addition <strong>of</strong> anabolic therapy, such as parathyroid<br />

hormone [48].<br />

S.H. Madsen et al. / Steroids 76 (2011) 1474–1482 1481<br />

75<br />

Acknowledgment<br />

This work was partly funded by a grant from the Danish Research<br />

Foundation, The Ministry <strong>of</strong> Science Technology and<br />

Innovation, Denmark (08-036109) with a stipend for the corresponding<br />

author.<br />

References<br />

[1] Engvall IL, Svensson B, Tengstrand B, Brismar K, Hafstrom I. Impact <strong>of</strong> low-dose<br />

prednisolone on <strong>bone</strong> synthesis and resorption in early rheumatoid arthritis:<br />

experiences from a two-year randomized study. Arthritis Res Ther<br />

2008;10(6):R128.<br />

[2] Hepper CT, Halvorson JJ, Duncan ST, Gregory AJ, Dunn WR, Spindler KP. The<br />

efficacy and duration <strong>of</strong> intra-articular corticosteroid injection for knee<br />

osteoarthritis: a systematic review <strong>of</strong> level I studies. J Am Acad Orthop Surg<br />

2009;17(10):638–46.<br />

[3] DiBattista JA, Martel-Pelletier J, Antakly T, Tardif G, Cloutier JM, Pelletier JP.<br />

Reduced expression <strong>of</strong> glucocorticoid receptor levels in human osteoarthritic<br />

chondrocytes. Role in the suppression <strong>of</strong> metalloprotease synthesis. J Clin<br />

Endocrinol Metab 1993;76(5):1128–34.<br />

[4] Chrysis D, Ritzen EM, Savendahl L. Growth retardation induced by<br />

dexamethasone is associated with increased apoptosis <strong>of</strong> the growth plate<br />

chondrocytes. J Endocrinol 2003;176(3):331–7.<br />

[5] Chrysis D, Zaman F, Chagin AS, Takigawa M, Savendahl L. Dexamethasone<br />

induces apoptosis in proliferative chondrocytes through activation <strong>of</strong> caspases<br />

and suppression <strong>of</strong> the Akt–phosphatidylinositol 3 0 -kinase signaling pathway.<br />

Endocrinology 2005;146(3):1391–7.<br />

[6] Sadowski T, Steinmeyer J. Effects <strong>of</strong> non-steroidal antiinflammatory drugs and<br />

dexamethasone on the activity and expression <strong>of</strong> matrix metalloproteinase-1,<br />

matrix metalloproteinase-3 and tissue inhibitor <strong>of</strong> metalloproteinases-1 by<br />

bovine articular chondrocytes. Osteoarthritis Cartilage 2001;9(5):407–15.<br />

[7] Nakazawa F, Matsuno H, Yudoh K, Watanabe Y, Katayama R, Kimura T.<br />

Corticosteroid treatment induces chondrocyte apoptosis in an experimental<br />

arthritis model and in chondrocyte cultures. Clin Exp Rheumatol<br />

2002;20(6):773–81.<br />

[8] Richardson DW, Dodge GR. Dose-dependent effects <strong>of</strong> corticosteroids on the<br />

expression <strong>of</strong> matrix-related genes in normal and cytokine-treated articular<br />

chondrocytes. Inflamm Res 2003;52(1):39–49.<br />

[9] Garvican ER, Vaughan-Thomas A, Redmond C, Gabriel N, Clegg PD. MMPmediated<br />

collagen breakdown induced by activated protein C in equine<br />

cartilage is reduced by corticosteroids. J Orthop Res 2010;28(3):370–8.<br />

[10] Eastell R, Reid DM, Compston J, Cooper C, Fogelman I, Francis RM, et al. A UK<br />

Consensus Group on management <strong>of</strong> glucocorticoid-induced osteoporosis: an<br />

update. J Intern Med 1998;244(4):271–92.<br />

[11] Hooyman JR, Melton III LJ, Nelson AM, O’Fallon WM, Riggs BL. Fractures after<br />

rheumatoid arthritis. A population-based study. Arthritis Rheum<br />

1984;27(12):1353–61.<br />

[12] Caplan L, Saag KG. Glucocorticoids and the risk <strong>of</strong> osteoporosis. Expert Opin<br />

Drug Saf 2009;8(1):33–47.<br />

[13] Canalis E, Mazziotti G, Giustina A, Bilezikian JP. Glucocorticoid-induced<br />

osteoporosis: pathophysiology and therapy. Osteoporos Int<br />

2007;18(10):1319–28.<br />

[14] H<strong>of</strong>bauer LC, Zeitz U, Schoppet M, Skalicky M, Schuler C, Stolina M, et al.<br />

Prevention <strong>of</strong> glucocorticoid-induced <strong>bone</strong> loss in mice by inhibition <strong>of</strong> RANKL.<br />

Arthritis Rheum 2009;60(5):1427–37.<br />

[15] Jia D, O’Brien CA, Stewart SA, Manolagas SC, Weinstein RS. Glucocorticoids act<br />

directly on osteoclasts to increase their life span and reduce <strong>bone</strong> density.<br />

Endocrinology 2006;147(12):5592–9.<br />

[16] Weinstein RS, Chen JR, Powers CC, Stewart SA, Landes RD, Bellido T, et al.<br />

Promotion <strong>of</strong> osteoclast survival and antagonism <strong>of</strong> bisphosphonate-induced<br />

osteoclast apoptosis by glucocorticoids. J Clin Invest 2002;109(8):1041–8.<br />

[17] Kim HJ, Zhao H, Kitaura H, Bhattacharyya S, Brewer JA, Muglia LJ, et al.<br />

Glucocorticoids suppress <strong>bone</strong> formation via the osteoclast. J Clin Invest<br />

2006;116(8):2152–60.<br />

[18] H<strong>of</strong>bauer LC, Gori F, Riggs BL, Lacey DL, Dunstan CR, Spelsberg TC, et al.<br />

Stimulation <strong>of</strong> osteoprotegerin ligand and inhibition <strong>of</strong> osteoprotegerin<br />

production by glucocorticoids in human osteoblastic lineage cells: potential<br />

paracrine mechanisms <strong>of</strong> glucocorticoid-induced osteoporosis. Endocrinology<br />

1999;140(10):4382–9.<br />

[19] Sivagurunathan S, Muir MM, Brennan TC, Seale JP, Mason RS. Influence <strong>of</strong><br />

glucocorticoids on human osteoclast generation and activity. J Bone Miner Res<br />

2005;20(3):390–8.<br />

[20] Oshina H, Sotome S, Yoshii T, Torigoe I, Sugata Y, Maehara H, et al. Effects <strong>of</strong><br />

continuous dexamethasone treatment on differentiation capabilities <strong>of</strong> <strong>bone</strong><br />

marrow-derived mesenchymal cells. Bone 2007;41(4):575–83.<br />

[21] Jager M, Fischer J, Dohrn W, Li X, Ayers DC, Czibere A, et al. Dexamethasone<br />

modulates BMP-2 effects on mesenchymal stem cells in vitro. J Orthop Res<br />

2008;26(11):1440–8.<br />

[22] Espina B, Liang M, Russell RG, Hulley PA. Regulation <strong>of</strong> bim in glucocorticoidmediated<br />

osteoblast apoptosis. J Cell Physiol 2008;215(2):488–96.


1482 S.H. Madsen et al. / Steroids 76 (2011) 1474–1482<br />

[23] Mak W, Shao X, Dunstan CR, Seibel MJ, Zhou H. Biphasic glucocorticoiddependent<br />

regulation <strong>of</strong> Wnt expression and its inhibitors in mature<br />

osteoblastic cells. Calcif Tissue Int 2009;85(6):538–45.<br />

[24] Buttgereit F, Zhou H, Kalak R, Gaber T, Spies CM, Huscher D, et al. Transgenic<br />

disruption <strong>of</strong> glucocorticoid signaling in mature osteoblasts and osteocytes<br />

attenuates K/BxN mouse serum-induced arthritis in vivo. Arthritis Rheum<br />

2009;60(7):1998–2007.<br />

[25] Zalavras C, Shah S, Birnbaum MJ, Frenkel B. Role <strong>of</strong> apoptosis in glucocorticoidinduced<br />

osteoporosis and osteonecrosis. Crit Rev Eukaryot Gene Expr<br />

2003;13(2–4):221–35.<br />

[26] Kerachian MA, Seguin C, Harvey EJ. Glucocorticoids in osteonecrosis <strong>of</strong> the<br />

femoral head: a new understanding <strong>of</strong> the mechanisms <strong>of</strong> action. J Steroid<br />

Biochem Mol Biol 2009;114(3–5):121–8.<br />

[27] Karsdal MA, Leeming DJ, Dam EB, Henriksen K, Alexandersen P, Pastoureau P,<br />

et al. Should <strong>subchondral</strong> <strong>bone</strong> turnover be targeted when treating<br />

osteoarthritis? Osteoarthritis Cartilage 2008;16(6):638–46.<br />

[28] Kwan TS, Lajeunesse D, Pelletier JP, Martel-Pelletier J. Targeting <strong>subchondral</strong><br />

<strong>bone</strong> for treating osteoarthritis: what is the evidence? Best Pract Res Clin<br />

Rheumatol 2010;24(1):51–70.<br />

[29] Madsen SH, Goettrup AS, Thomsen G, Christensen ST, Schultz N, Henriksen K,<br />

et al. Characterization <strong>of</strong> an ex vivo femoral head model assessed by markers<br />

<strong>of</strong> <strong>bone</strong> and cartilage turnover. Cartilage 2011;2(3):265–78.<br />

[30] Karsdal MA, Madsen SH, Christiansen C, Henriksen K, Fosang AJ, Sondergaard<br />

BC. Cartilage degradation is fully reversible in the presence <strong>of</strong> aggrecanase but<br />

not matrix metalloproteinase activity. Arthritis Res Ther 2008;10(3):R63.<br />

[31] Sorensen MG, Henriksen K, Schaller S, Henriksen DB, Nielsen FC, Dziegiel MH,<br />

et al. Characterization <strong>of</strong> osteoclasts derived from CD14 + monocytes isolated<br />

from peripheral blood. J Bone Miner Metab 2007;25(1):36–45.<br />

[32] Ghosh-Choudhury N, Windle JJ, Koop BA, Harris MA, Guerrero DL, Wozney JM,<br />

et al. Immortalized murine osteoblasts derived from BMP 2-T-antigen<br />

expressing transgenic mice. Endocrinology 1996;137(1):331–9.<br />

[33] Olsen AK, Sondergaard BC, Byrjalsen I, Tanko LB, Christiansen C, Muller A, et al.<br />

Anabolic and catabolic function <strong>of</strong> chondrocyte ex vivo is reflected by the<br />

metabolic processing <strong>of</strong> type II collagen. Osteoarthritis Cartilage<br />

2007;15(3):335–42.<br />

[34] Karsdal MA, Neutzsky-Wulff AV, Dziegiel MH, Christiansen C, Henriksen K.<br />

Osteoclasts secrete non-<strong>bone</strong> derived signals that induce <strong>bone</strong> formation.<br />

Biochem Biophys Res Commun 2008;366(2):483–8.<br />

[35] Jonsson KB, Frost A, Larsson R, Ljunghall S, Ljunggren O. A new fluorometric<br />

assay for determination <strong>of</strong> osteoblastic proliferation: effects <strong>of</strong> glucocorticoids<br />

and insulin-like growth factor-I. Calcif Tissue Int 1997;60(1):30–6.<br />

[36] Bay-Jensen AC, Liu Q, Byrjalsen I, Li Y, Wang J, Pedersen C, et al. Enzyme-linked<br />

immunosorbent assay (ELISAs) for metalloproteinase derived type II collagen<br />

neoepitope, CIIM-increased serum CIIM in subjects with severe radiographic<br />

osteoarthritis. Clin Biochem 2011;44(5-6):423–9.<br />

[37] Sumer EU, Sondergaard BC, Rousseau JC, Delmas PD, Fosang AJ, Karsdal MA,<br />

et al. MMP and non-MMP-mediated release <strong>of</strong> aggrecan and its fragments<br />

from articular cartilage: a comparative study <strong>of</strong> three different aggrecan and<br />

glycosaminoglycan assays. Osteoarthritis Cartilage 2007;15(2):212–21.<br />

[38] Henriksen K, Gram J, Neutzsky-Wulff AV, Jensen VK, Dziegiel MH, Bollerslev J,<br />

et al. Characterization <strong>of</strong> acid flux in osteoclasts from patients harboring a<br />

G215R mutation in ClC-7. Biochem Biophys Res Commun 2009;378(4):804–9.<br />

76<br />

[39] Camplejohn KL, Allard SA. Limitations <strong>of</strong> safranin ‘O’ staining in proteoglycandepleted<br />

cartilage demonstrated with monoclonal antibodies. Histochemistry<br />

1988;89(2):185–8.<br />

[40] Habib GS. Systemic effects <strong>of</strong> intra-articular corticosteroids. Clin Rheumatol<br />

2009;28(7):749–56.<br />

[41] Skwara A, Peterlein CD, Tibesku CO, Rosenbaum D, Fuchs-Winkelmann S.<br />

Changes <strong>of</strong> gait patterns and muscle activity after intraarticular treatment <strong>of</strong><br />

patients with osteoarthritis <strong>of</strong> the knee: a prospective, randomised,<br />

doubleblind study. Knee 2009;16(6):466–72.<br />

[42] Mansell JP, Collins C, Bailey AJ. Bone, not cartilage, should be the major focus in<br />

osteoarthritis. Nat Clin Pract Rheumatol 2007;3(6):306–7.<br />

[43] van der Kraan PM, Vitters EL, Postma NS, Verbunt J, Van den Berg WB.<br />

Maintenance <strong>of</strong> the synthesis <strong>of</strong> large proteoglycans in anatomically intact<br />

murine articular cartilage by steroids and insulin-like growth factor I. Ann<br />

Rheum Dis 1993;52(10):734–41.<br />

[44] Madsen SH, Sumer EU, Bay-Jensen AC, Sondergaard BC, Qvist P, Karsdal MA.<br />

Aggrecanase- and matrix metalloproteinase-mediated aggrecan degradation is<br />

associated with different molecular characteristics <strong>of</strong> aggrecan and separated<br />

in time ex vivo. Biomarkers 2010;15(3):266–76.<br />

[45] Augustine AJ, Oleksyszyn J. Glucocorticosteroids inhibit degradation in bovine<br />

cartilage explants stimulated with concomitant plasminogen and interleukin-<br />

1 alpha. Inflamm Res 1997;46(2):60–4.<br />

[46] Rauch A, Seitz S, Baschant U, Schilling AF, Illing A, Stride B, et al.<br />

Glucocorticoids suppress <strong>bone</strong> formation by attenuating osteoblast<br />

differentiation via the monomeric glucocorticoid receptor. Cell Metab<br />

2010;11(6):517–31.<br />

[47] Kalak R, Zhou H, Street J, Day RE, Modzelewski JR, Spies CM, et al. Endogenous<br />

glucocorticoid signalling in osteoblasts is necessary to maintain normal <strong>bone</strong><br />

structure in mice. Bone 2009;45(1):61–7.<br />

[48] Weinstein RS, Jilka RL, Almeida M, Roberson PK, Manolagas SC. Intermittent<br />

parathyroid hormone administration counteracts the adverse effects <strong>of</strong><br />

glucocorticoids on osteoblast and osteocyte viability, <strong>bone</strong> formation, and<br />

strength in mice. Endocrinology 2010;151(6):2641–9.<br />

[49] Henriksen K, Neutzsky-Wulff AV, Bonewald LF, Karsdal MA. Local<br />

communication on and within <strong>bone</strong> controls <strong>bone</strong> remodeling. Bone<br />

2009;44(6):1026–33.<br />

[50] Jevon M, Hirayama T, Brown MA, Wass JA, Sabokbar A, Ostelere S, et al.<br />

Osteoclast formation from circulating precursors in osteoporosis. Scand J<br />

Rheumatol 2003;32(2):95–100.<br />

[51] Takuma A, Kaneda T, Sato T, Ninomiya S, Kumegawa M, Hakeda Y.<br />

Dexamethasone enhances osteoclast formation synergistically with<br />

transforming growth factor-beta by stimulating the priming <strong>of</strong> osteoclast<br />

progenitors for differentiation into osteoclasts. J Biol Chem<br />

2003;278(45):44667–74.<br />

[52] Soe K, Delaisse JM. Glucocorticoids maintain human osteoclasts in the active<br />

mode <strong>of</strong> their resorption cycle. J Bone Miner Res 2010 [Epub ahead <strong>of</strong> print].<br />

Ref Type: Generic.


CHAPTER 6<br />

CHAPTER 6: PAPER III<br />

PAPER III<br />

___________________________________________________________________________<br />

Introductory remarks<br />

This manuscript focuses on the pathogenesis <strong>of</strong> cartilage, and evaluates the different proteolytic<br />

degradation processes for aggrecan, which vary <strong>between</strong> the OA-stages. It is <strong>important</strong> to<br />

understand the different degrading processes <strong>of</strong> aggrecan if we want to target the right proteases<br />

at a certain stage <strong>of</strong> disease.<br />

77


Biomarkers Downloaded from informahealthcare.com by Panum Biblioteket on 08/31/10<br />

For personal use only.<br />

Biomarkers, 2010; 15(3): 266–276<br />

original article<br />

Aggrecanase- and matrix metalloproteinase- mediated<br />

aggrecan degradation is associated with different<br />

molecular characteristics <strong>of</strong> aggrecan and separated in<br />

time ex vivo<br />

Suzi Hoegh Madsen, Eren Ufuk Sumer, Anne-Christine Bay-Jensen, Bodil-Cecilie Sondergaard,<br />

Per Qvist, and Morten Asser Karsdal<br />

Nordic Bioscience A/S, Herlev, Denmark<br />

Introduction<br />

abstract<br />

Aggrecan is one <strong>of</strong> the first proteins to be depleted from articular cartilage in early osteoarthritis. We investigated<br />

the molecular differences <strong>between</strong> matrix metalloproteinase (MMP)- and aggrecanase-mediated<br />

aggrecan degradation, as a consequence <strong>of</strong> their distinct time-dependent degradation pr<strong>of</strong>iles. Cartilage<br />

degradation was induced by cytokine stimulation in bovine articular cartilage explants and quantified by<br />

a dye-binding assay and immunoassays. The size <strong>of</strong> degradation fragments was analysed by Western blot.<br />

Cytokine stimulation resulted in the early release <strong>of</strong> aggrecanase-mediated aggrecan degradation fragments.<br />

In contrast, MMP-mediated aggrecan degradation began only at day 16 and continued to day 21.<br />

Western blot analysis showed that glycosylated high-molecular-weight 374 ARGSVI fragments appeared at<br />

day 7, in contrast to deglycosylated low-molecular-weight 342 FFGVG fragments which were detected at day<br />

21. Aggrecan degradation may be divided into two different pools, a high-molecular-weight aggrecanasemediated<br />

pool, and a low-molecular-weight MMP-mediated pool. This may have implications for the development<br />

<strong>of</strong> intervention strategies for OA.<br />

Keywords: Proteoglycans; cartilage; osteoarthritis; ELISA; biomarkers; aggrecanases; MMPs<br />

Cartilage is normally maintained by a balance <strong>between</strong><br />

catabolic and anabolic processes. However, in joint<br />

degenerative diseases, such as osteoarthritis (OA), the<br />

rate <strong>of</strong> cartilage degradation exceeds the rate <strong>of</strong> formation,<br />

resulting in a net loss <strong>of</strong> cartilage matrix (Behrens<br />

et al. 1989, Felson & Neogi 2004). The extracellular matrix<br />

(ECM) consists mainly <strong>of</strong> collagen type II, which forms<br />

a fibrous network that entraps proteoglycans <strong>of</strong> which<br />

aggrecan is the most predominant (Hardingham &<br />

Fosang 1995, Kiani et al. 2002).<br />

Aggrecan is one <strong>of</strong> the ECM proteins to be degraded<br />

in OA (Caterson et al. 2000), primarily by ADAMTSs<br />

(a disintegrin and metalloproteinase domain with<br />

thrombospondin motifs) (Glasson et al. 2005, Struglics et al.<br />

2006) and matrix metalloproteinases (MMPs) (Fosang et al.<br />

1996, Lark et al. 1997, Struglics et al. 2006). Aggrecanase<br />

activity leads to the release <strong>of</strong> glycosaminoglycans (GAGs),<br />

although it is not known if MMPs also play a role in aggrecan<br />

GAG loss (Sandy 2006, Struglics et al. 2006). As a consequence<br />

<strong>of</strong> aggrecan loss, the cartilage exhibits altered<br />

matrix properties such as loss <strong>of</strong> fixed negatively charged<br />

density <strong>of</strong> the tissue and thereby lower water content and<br />

elasticity (Roughley & Lee 1994). A deeper understanding<br />

<strong>of</strong> the molecular mechanisms involved in proteoglycan<br />

depletion may aid the development <strong>of</strong> efficient diseasemodifying<br />

osteoarthritic drugs (DMOADs).<br />

The core protein <strong>of</strong> aggrecan has three globular<br />

domains: G1, G2 and G3. G1 and G2 are separated by<br />

Address for Correspondence: Suzi Hoegh Madsen, Nordic Bioscience A/S, Herlev Hovedgade 207, DK-2730 Herlev, Denmark. Tel: + 45 44 52 52 52. Fax:<br />

+ 45 44 52 52 51. E-mail: shm@nordicbioscience.com<br />

(Received 03 August 2009; revised 01 December 2009; accepted 01 December 2009)<br />

ISSN 1354-750X print/ISSN 1366-5804 online © 2010 Informa UK Ltd<br />

DOI: 10.3109/13547500903521810 http://www.informahealthcare.com/bmk<br />

78


Biomarkers Downloaded from informahealthcare.com by Panum Biblioteket on 08/31/10<br />

For personal use only.<br />

the interglobular domain (IGD), which is highly sensitive<br />

to proteolysis (Flannery et al. 1992, Hardingham<br />

& Fosang 1995). G2 and G3 are heavily saturated with<br />

highly negatively charged GAGs (Flannery et al. 1992,<br />

Hardingham & Fosang 1995) (Figure 1). The G2 domain<br />

is unique to aggrecan, and sandwich enzyme-linked<br />

immunosorbent assays (ELISAs) <strong>of</strong> aggrecan degradation<br />

have been developed against peptides with the G2<br />

and the neoepitopes 342 FFGVG or 374 ARGSV (Karsdal<br />

et al. 2007, 2008) generated by MMPs (Flannery et al.<br />

1992) and ADAMTS (Tortorella et al. 1999), respectively.<br />

The ADAMTS-specific 374 ARGSV site is located <strong>between</strong><br />

the 342 FFGVG site and the G2 domain. The 342 FFGVG-G2<br />

fragments assayed in the present study represent only<br />

MMP-mediated degradation, which has not already<br />

been processed by ADAMTSs. The 374 ARGSV-G2 represents<br />

fragments, which theoretically may have been<br />

pre-processed by MMPs at the 342 FFGVG site (Figure 1),<br />

although the available evidence suggests that this is very<br />

unlikely (Mercuri et al. 2000).<br />

Studies with ex vivo cultures have shown that cytokinestimulated<br />

cartilage releases collagen and proteoglycan<br />

fragments to the conditioned medium (Sondergaard<br />

et al. 2006a). These fragments can be detected by novel<br />

ELISA assays, which measure neoepitopes cleaved by<br />

specific proteases. These degradation patterns resemble<br />

the molecular mechanisms seen in the pathogenesis<br />

<strong>of</strong> OA (Caterson et al. 2000). Our group has previously<br />

shown that cytokine-stimulated cartilage releases products<br />

with specific neoepitopes from collagen type II and<br />

aggrecan (Karsdal et al. 2007, 2008, Rousseau et al. 2008,<br />

G1<br />

MMP<br />

IPEN 341 342 FFGV<br />

AF-28<br />

ADAMTS<br />

Aggrecanase- and MMP-mediated aggrecan degradation 267<br />

IGD sGAG<br />

TEGE 373 374 ARGS<br />

BC-3<br />

Sondergaard 2006a). Furthermore, we also showed that<br />

MMP-2 and -9 activity increased over time in cytokinestimulated<br />

cartilage explants (Sondergaard 2006a),<br />

which is <strong>important</strong> when investigating the difference<br />

<strong>between</strong> aggrecanase- and MMP-mediated degradation<br />

<strong>of</strong> aggrecan.<br />

Different research groups have suggested that articular<br />

cartilage aggrecan may be present in the tissue in<br />

two or more metabolic pools (Ilic et al. 1995, Lark et al.<br />

1997, Lohmander et al. 1973, Mok et al. 1994, Struglics<br />

et al. 2006). However, currently, the temporal and spatial<br />

molecular degradation <strong>of</strong> aggrecan has not been<br />

clarified, and various contradictory theories and models<br />

have been proposed by different investigators (Fosang<br />

et al. 2000, Lark et al. 1997, Sandy 2006, Struglics et al.<br />

2006). It has been proposed by Fosang et al. (2000) that<br />

aggrecan degradation by ADAMTSs and MMPs occurs in<br />

independent locations and with different kinetics. More<br />

recently Struglics et al. (2006) analysed human synovial<br />

fluids and concluded that ADAMTSs and MMPs act<br />

sequentially on the same substrate molecule. A review<br />

<strong>of</strong> the area by Sandy (2006) attempted to clarify these<br />

controversies. On the basis <strong>of</strong> available evidence, Sandy<br />

(2006) concluded that ADAMTSs and MMPs degrade<br />

different aggrecan structures. Specifically it was suggested<br />

that ADAMTSs act primarily on full-length or<br />

G1-G2-chondroitin sulfate1 aggrecan by a sequential<br />

process which removes the chondroitin sulfate-bearing<br />

domains and lastly at the Glu 373-374 Ala bond in the IGD.<br />

In contrast it was proposed that MMPs degrade only the<br />

calpain (calcium-dependent cysteine protease) product<br />

G2 G3<br />

F-78-HRP<br />

F-78-HRP<br />

Figure 1. Protease-mediated neoepitopes in aggrecan. Aggrecan molecule with three globular domains G1, G2 and G3. The interglobular domain<br />

(IGD) is highly sensitive to proteolysis. Beneath the aggrecan molecule are the 342 FFGVG-G2 and 374 ARGSV-G2 fragments mediated by matrix metalloproteinase<br />

(MMP) and ADAMTS, respectively. The antibodies BC-3 and AF-28 recognize the neoepitopes and F-78 is the detection antibody<br />

binding to the G2 domain. The domain <strong>between</strong> G2 and G3 is heavily saturated with highly negatively charged glycosaminoglycans (GAGs).<br />

79


Biomarkers Downloaded from informahealthcare.com by Panum Biblioteket on 08/31/10<br />

For personal use only.<br />

268 Suzi Hoegh Madsen et al.<br />

G1-G2-keratan sulfate (Maehara et al. 2007, Oshita et al.<br />

2004) by cleavage at the 342 FFGVG site. In this scenario<br />

the independent activities <strong>of</strong> ADAMTSs and MMPs are<br />

seen as being due to a protease-specific preference for<br />

different substrate structures. Furthermore, Lark et al.<br />

(1997) described three hypothetical models for pathways<br />

<strong>of</strong> proteolytic degradation <strong>of</strong> aggrecan, emphasizing the<br />

complex nature <strong>of</strong> the generation <strong>of</strong> various fragments<br />

from the same molecule.<br />

To examine further the kinetics and molecular<br />

dynamics <strong>of</strong> aggrecanolysis in cartilage explant cultures,<br />

we investigated the degradation process by quantifying<br />

the time-dependent release <strong>of</strong> ADAMTS- and MMPmediated<br />

aggrecan fragments by the 374 ARGSVI-G2<br />

and 342 FFGVG-G2 ELISA assays, respectively. These<br />

new aggrecan fragments were categorized by size, via<br />

Western blot, to identify molecular differences regarding<br />

glycosylation <strong>of</strong> the core protein. Furthermore, we investigated<br />

the specificity pattern <strong>of</strong> MMP-9 and ADAMTS-4<br />

for degrading native and deglycosylated aggrecan in<br />

vitro with respect to generating the aforementioned ex<br />

vivo neoepitopes.<br />

Materials and methods<br />

Tissue preparation<br />

Bovine articular cartilage explants were harvested by<br />

using a scalpel to remove the articular cartilage without<br />

adherent calcified cartilage from stifle joints <strong>of</strong><br />

18-month-old bovine heifers. The cartilage explants<br />

(12 ± 2 mg) were washed in phosphate-buffered saline<br />

(PBS) and cultured in 96-well plates (Nunc, Slangerup,<br />

Denmark) in four replicas with 200 µl serum-free<br />

medium (Life Technologies, Naerum, Denmark) plus<br />

penicillin/streptomycin (Invitrogen, Taastrup, Denmark)<br />

per well in the absence or presence <strong>of</strong> tumour necrosis<br />

factor (TNF)-α (10 ng ml −1 ) (R&D Systems, Abingdon,<br />

UK) and oncostatin M (OSM) (20 ng ml −1 ) (Sigma<br />

Aldrich, Broendby, Denmark). These concentrations<br />

had been optimized in previous ex vivo model studies<br />

by Sondergaard et al. (2006a, b). As a negative control,<br />

cartilage was metabolically inactivated (MI) by three<br />

repeated freeze–thaw cycles in liquid N 2 , and at 37°C in<br />

a water bath. The explants culture media were replaced<br />

every 2nd or 3rd day for 21 days. The conditioned media<br />

were kept at -20°C until tested for the presence <strong>of</strong> biochemical<br />

markers.<br />

Biochemical markers <strong>of</strong> aggrecan degradation<br />

MMP-mediated aggrecan degradation was quantified<br />

using the 342 FFGVG-G2 sandwich ELISA assay (Sumer<br />

80<br />

et al. 2007), with AF-28 (Fosang et al. 1995) as the<br />

catching antibody. Quantification <strong>of</strong> the aggrecanasemediated<br />

374 ARGSV-G2 neoepitope was obtained in<br />

a similar way to the 342 FFGVG-G2 ELISA, except the<br />

catching antibody was BC-3 (Abcam, Cambridge, UK)<br />

to recognize the aggrecanase-mediated neoepitope<br />

374 ARGSVI (Karsdal 2007). Both assays required the presence<br />

<strong>of</strong> the G2 domain on the fragments being assayed,<br />

with the detecting antibody being F-78. The efficacy <strong>of</strong><br />

the antibodies was confirmed by blocking experiments<br />

in cytokine-stimulated explant media with the peptides<br />

342 FFGVGEEDITVQT and 374 ARGSVILTVKPIF (Bachem,<br />

Weil am Rhein, Germany).<br />

Detection <strong>of</strong> sulfated glycosaminoglycans<br />

Sulfated glycosaminoglycan (sGAG) levels were measured<br />

in the conditioned medium using the alcian blue-binding<br />

assay according to the manufacturer’s instructions (Euro-<br />

Diagnostica, Malmö, Sweden).<br />

Western blot on supernatants<br />

Conditioned medium from four replicates <strong>of</strong> MI, nontreated,<br />

or cytokine-stimulated cartilage explants from<br />

day 7 or 21 were either deglycosylated for 4 h with keratanase<br />

and chondroitinase lyase ABC (0.1 units/10 μg)<br />

(Sigma Aldrich) with gentle shaking at 37°C in 100 mM<br />

Tris/HCl and sodium acetate buffer at pH 6.5, or left<br />

untreated. Thirty microlitres <strong>of</strong> the deglycosylated or<br />

untreated sample was electrophoresed on 3–8% precasted<br />

Tris–acetate gradient gels under reducing conditions<br />

using Tris–acetate as running buffer (Invitrogen).<br />

After transferring the proteins to polyvinyl difluoride<br />

membranes overnight at room temperature (RT) at 120<br />

mA in a 10 mM N-cyclohexyl-3-aminopropanesulfonic<br />

acid buffer with 5% ethanol, the membranes were<br />

blocked with 50 ml <strong>of</strong> 5% non-fat milk powder, 0.2%<br />

Tween-20 in PBS with shaking for 2 h at RT. Antibodies<br />

were applied in appropriate dilutions in 50 ml PBS<br />

with 5% bovine non-fat milk, 0.2% Tween-20 for 1 h at<br />

RT. BC-3 anti- 374 ARGSVI cell supernatants (Caterson<br />

et al. 2000) diluted 1:50 or the anti- 342 FFGVG monoclonal<br />

antibody AF-28 diluted 1:2250, giving a final<br />

concentration <strong>of</strong> 1200 ng ml −1 IgG (Fosang et al. 1995).<br />

After washing three times with PBS containing 0.2%<br />

Tween-20, the membranes were incubated for 1 h at RT<br />

with peroxidase-labelled rabbit antimouse antibody<br />

(1:5000) (Millipore, Copenhagen, Denmark) in PBS<br />

with 5% bovine non-fat milk powder, 0.2% Tween-20.<br />

After washing three times with PBS (0.2% Tween-20),<br />

the bands were visualized using an electrochemiluminescence<br />

developer system (Amersham, Hilleroed,<br />

Denmark).


Biomarkers Downloaded from informahealthcare.com by Panum Biblioteket on 08/31/10<br />

For personal use only.<br />

ELISA for determining the total aggrecanase activity<br />

The total aggrecanase activity in the conditioned medium<br />

from days 7–21 was measured in four replicates using a<br />

commercially available ELISA assay according to the<br />

manufacturer’s instructions (MD Bioscience, Zurich,<br />

Switzerland).<br />

Digestion <strong>of</strong> aggrecan fragments in the conditioned<br />

medium with ADAMTS-4<br />

The conditioned medium from cytokine-stimulated<br />

cartilage explants from day 21 was diluted 1:50 in<br />

aggrecanase digestion buffer (50 mM Tris–HCI, pH 7.5,<br />

150 mM NaCl, 5 mM CaCl 2 , 1 μM leupeptin, 1 μM pepstatin,<br />

1mM Pefabloc, 0.05% Tween-20) and digested for<br />

15 min with shaking at 37°C, using a total concentration<br />

<strong>of</strong> 20 nM <strong>of</strong> ADAMTS-4 (Millipore). The digestions were<br />

stopped with 0.5 mM EDTA and the media tested using<br />

the 342 FFGVG-G2 and 374 ARGSV-G2 assays.<br />

MMP-9 and ADAMTS-4 digestion on native and<br />

deglycosylated aggrecan<br />

Purified bovine aggrecan (Sigma Aldrich) was dissolved in<br />

chondroitinase buffer (50 mM Tris, pH 8, 60 mM sodium<br />

acetate, 0.02% BSA) to a concentration <strong>of</strong> 2 mg ml −1 . One<br />

half <strong>of</strong> the solution was deglycosylated with chondroitinase<br />

ABC (0.01 U/10 µg) for 4 h at 37°C with shaking.<br />

The other half was left untreated but incubated along<br />

the deglycosylated half. Two hours before terminating<br />

the deglycosylation, the MMP-9 enzyme (Calbiochem,<br />

Herlev, Denmark) was activated in 4-aminophenylmercuric<br />

acetate (APMA) (1 mM) at 37°C for 2 h. The native<br />

and deglycosylated aggrecan were each digested with<br />

MMP-9 (1:100 enzyme/protein) and ADAMTS-4 (1:50<br />

enzyme/protein). Digestion with MMP-9 continued for<br />

3 days in a MMP buffer (100 mM Tris–HCl, 100 mM NaCl,<br />

10 mM CaCl 2 , 2 mM Zn-acetate, pH 8) while digestion<br />

with ADAMTS-4 took place over 22 h in a ADAMTS-4<br />

buffer (75 mM Tris–HCl, 5 mM CaCl 2 .2H 2 O, 150 mM NaCl,<br />

pH 7.5) at 37°C, and was stopped with 0.5 mM EDTA. The<br />

MMP-9 and ADAMTS-4 digestions on native and deglycosylated<br />

aggrecan were examined using the 342 FFGVG-G2<br />

and 374 ARGSV-G2 ELISA assays.<br />

Statistics<br />

Results are shown as mean + standard error <strong>of</strong> mean<br />

(SEM). Baseline was set at day 2 <strong>of</strong> culturing. Differences<br />

<strong>between</strong> mean values were compared using the Student’s<br />

t-test for unpaired observations, assuming normal distribution<br />

where more than four replicates were used.<br />

Differences were considered statistically significant if<br />

p < 0.05.<br />

Aggrecanase- and MMP-mediated aggrecan degradation 269<br />

81<br />

Results<br />

Depletion <strong>of</strong> sulfated glycosaminoglycans in<br />

cytokine-stimulated cartilage explants is mediated by<br />

aggrecanases<br />

As aggrecan is one <strong>of</strong> the first proteins to deplete from<br />

articular cartilage in early OA, we investigated the timedependent<br />

enzyme-mediated release pattern <strong>of</strong> aggrecan<br />

molecules from cytokine-stimulated bovine articular<br />

cartilage explants. The first cytokine-stimulated release<br />

<strong>of</strong> aggrecan fragments from bovine articular cartilage<br />

explants was caused by aggrecanase activity, as demonstrated<br />

by an increase in fragments detected by the<br />

374 ARGSV-G2 assay. The maximum level <strong>of</strong> aggrecan fragments<br />

was detected at day 7 (Figure 2A) corresponding<br />

to an approximate sevenfold increase compared with<br />

non-stimulated bovine articular cartilage explants. After<br />

the peak at day 7, 374 ARGSVI-G2 concentrations were<br />

reduced to background levels.<br />

To investigate if the reduction in 374 ARGSVI-G2 after<br />

day 7 resulted from depletion <strong>of</strong> the aggrecan protein as a<br />

substrate, we measured the MMP-mediated 342 FFGVG-G2<br />

fragments. A time-dependent analysis showed that after<br />

day 16, increased 342 FFGVG-G2 fragments were released<br />

from cytokine-stimulated cartilage. At day 21, an increase<br />

<strong>of</strong> more than 1600% in these released fragments was<br />

observed in cytokine-stimulated cartilage compared<br />

with non-stimulated cartilage (Figure 2B). This indicted<br />

that aggrecan was present as a substrate during the entire<br />

culture period.<br />

Karsdal et al. (2008) showed a total depletion <strong>of</strong> sGAGs,<br />

assessed by alcian blue staining from bovine cartilage<br />

explants as early as after 7 days after cytokine stimulation,<br />

while the non-stimulated cartilage retained the sGAGs in<br />

the ECM. Measurements <strong>of</strong> the time-dependent release<br />

<strong>of</strong> sGAGs to the conditioned medium (Figure 2C) showed<br />

a similar pattern to the aggrecanase-mediated aggrecan<br />

degradation. This indicated that the loss <strong>of</strong> sGAGs may<br />

be linked to the loss <strong>of</strong> aggrecanase-mediated aggrecan<br />

degradation.<br />

Aggrecanase-mediated aggrecan degradation<br />

fragments are predominantly glycosylated whereas<br />

MMP-mediated aggrecan degradation fragments are<br />

predominantly deglycosylated<br />

Due to the strong correlation <strong>between</strong> aggrecanasemediated<br />

aggrecan degradation and sGAG-substituted<br />

molecules, we investigated aggrecan fragments in the<br />

early period (day 7) and late period (day 21) by Western<br />

blotting.<br />

In the conditioned medium from cytokine-stimulated<br />

cartilage from day 7, a high-molecular-weight band –<br />

above 460 kDa – <strong>of</strong> the aggrecanase-mediated 374 ARGSVI


Biomarkers Downloaded from informahealthcare.com by Panum Biblioteket on 08/31/10<br />

For personal use only.<br />

270 Suzi Hoegh Madsen et al.<br />

A<br />

µg 374ARGSVI-G2/ml/ mg cartilage<br />

B<br />

µg 342FFGVG-G2/ml/ mg cartilage<br />

C<br />

µg GAGs/ml/<br />

mg cartilage<br />

125<br />

100<br />

10<br />

75<br />

50<br />

25<br />

8<br />

6<br />

4<br />

2<br />

0<br />

60<br />

40<br />

20<br />

0<br />

0<br />

**<br />

*<br />

2<br />

*<br />

*<br />

Days<br />

Cytokines<br />

W/O<br />

neoepitope was recognized by the monoclonal antibody<br />

BC-3 (Figure 3A). This band was reduced to approximately<br />

325 kDa upon deglycosylation (Figure 3B). In<br />

contrast, the low-molecular-weight smear <strong>of</strong> bands<br />

carrying the 342 FFGVG neoepitopes detected in the<br />

supernatant at day 21 with monoclonal antibody AF-28<br />

*<br />

4 7 9 11 1416 18 21<br />

Days<br />

* **<br />

* **<br />

2 4 7 9 11 14 16 18 21<br />

**<br />

**<br />

2 4 7 9 11 14 16 18 21<br />

Days<br />

MI<br />

Cytokines<br />

W/O<br />

MI<br />

Cytokines<br />

W/O<br />

Figure 2. Quantification <strong>of</strong> biochemical markers. Bovine articular<br />

cartilage explants were isolated and cultured with the proinflammatory<br />

cytokines oncostatin M (OSM) (10 ng ml −1 ) in combination with<br />

tumour necrosis factor (TNF)-α (20 ng ml −1 ) (- ◆ -). The time-dependent<br />

release <strong>of</strong> aggrecanase-mediated 374 ARGSV-G2 (A), matrix metalloproteinase<br />

(MMP)-mediated 342 FFGVG-G2 fragments (B) or sulfated<br />

glycosaminoglycans (GAGs) (C) were quantified. Non-stimulated<br />

explants (- ■ -) were used as the vehicle. Metabolically inactivated<br />

(MI) explants (- ● -) were used as a negative control to measure the<br />

non-chondrocyte-mediated release. The asterisks indicate significant<br />

difference compared with non-treated explants; *p < 0.05, **p < 0.01,<br />

***p < 0.001; n = 4. W/O, non-stimulated controls).<br />

MI<br />

82<br />

(Figure 3C) was relatively insensitive to deglycosylation<br />

(Figure 3D). No bands were detected when BC-3 was<br />

used for staining <strong>of</strong> the conditioned media from day 21,<br />

or the monoclonal antibody AF-28 from day 7 (data not<br />

shown), which resemble the time-dependent analysis by<br />

the 374 ARGSV-G2 and 342 FFGVG-G2 assays in Figure 2A<br />

and B.<br />

The calculated theoretic molecular weight difference<br />

<strong>between</strong> the neoepitopes 342 FFGVG-G2 and 374 ARGSV-G2<br />

is approximately 3.6 KDa (32 aa). However, the molecular<br />

weight difference <strong>between</strong> these two neoepitopes when<br />

released as fragments to the conditioned media was far<br />

from this. The 374 ARGSV-G2 fragment was glycosylated<br />

and large, and the 342 FFGVG-G2 fragment was deglycosylated<br />

and small. This explains the correlation <strong>between</strong><br />

the release patterns <strong>of</strong> sGAGs and aggrecanase-mediated<br />

aggrecan degradation. Furthermore, the Western blots<br />

clearly showed that the deglycosylated 374 ARGSV-G2<br />

fragments were more than 200 KDa larger than the<br />

deglycosylated 342 FFGVG-G2 fragments. This indicates<br />

that we were not able to deglycosylate the 374 ARGSV-G2<br />

fragments totally, and that the protein structure <strong>of</strong> the<br />

aggrecanase-mediated aggrecan fragments may be larger<br />

than the MMP-mediated 342 FFGVG-G2 fragments.<br />

Lack <strong>of</strong> aggrecanase-mediated aggrecan degradation<br />

is not a consequence <strong>of</strong> lack <strong>of</strong> aggrecanase activity<br />

In the late culture period, after day 16, we saw high<br />

levels <strong>of</strong> 342 FFGVG-G2 fragments and very low levels<br />

<strong>of</strong> 374 ARGSV-G2 fragments. To investigate if the lack <strong>of</strong><br />

aggrecanase-mediated aggrecan degradation was caused<br />

by depletion <strong>of</strong> aggrecanase activity, we tested the conditioned<br />

medium at days 7 and 21. The concentration <strong>of</strong><br />

aggrecanases released at day 7 from cytokines-stimulated<br />

cartilage did not show any increase compared with that<br />

from the non-stimulated cartilage. In contrast, the concentration<br />

<strong>of</strong> aggrecanases released at day 21 from cytokinestimulated<br />

cartilage revealed an elevation <strong>of</strong> over 935%<br />

compared with non-stimulated cartilage (Figure 4). This<br />

reduced release <strong>of</strong> aggrecanases at day 7 is interesting,<br />

because <strong>of</strong> the high aggrecanase-mediated degradation<br />

<strong>of</strong> aggrecan at the same time. However, the aggrecanases<br />

might still be sited in the extracellular matrix, which is<br />

then released later due to the loss <strong>of</strong> sGAGs. It appeared<br />

that the low concentration <strong>of</strong> 374 ARGSV-G2 fragments at<br />

day 21 was not due to low aggrecanase activity.<br />

The MMP-mediated deglycosylated aggrecan<br />

degradation released in the late period is not<br />

protected from further cleavage at the major<br />

aggrecanase site in the IGD in vitro<br />

Aggrecanases were present in the conditioned medium<br />

at the end <strong>of</strong> the culture period. To investigate whether


Biomarkers Downloaded from informahealthcare.com by Panum Biblioteket on 08/31/10<br />

For personal use only.<br />

Day 7<br />

Day 21<br />

the lack <strong>of</strong> 374 ARGSV-G2 fragments at day 21 was due to<br />

the inability <strong>of</strong> aggrecanases to process the 342 FFGVG-G2<br />

fragments further, we added recombinant enzymes to<br />

the culture medium and measured further cleavage <strong>of</strong><br />

the 342 FFGVG-G2 aggrecan fragments by quantitative<br />

measurements.<br />

A<br />

W/O MI<br />

C<br />

W/O<br />

MI<br />

Aggrecanase- and MMP-mediated aggrecan degradation 271<br />

Untreated Deglycosylated<br />

Cytokines<br />

Cytokines<br />

374 ARGSV<br />

460 kDa<br />

75 kDa<br />

342 FFGVG<br />

50 kDa<br />

B<br />

W/O<br />

D<br />

W/O<br />

MI Cytokines<br />

MI Cytokines<br />

460 kDa<br />

374 ARGSV<br />

268 kDa<br />

75 kDa<br />

342 FFGVG<br />

50 kDa<br />

Figure 3. Western blot analysis <strong>of</strong> aggrecanase- and matrix metalloproteinase (MMP)-mediated fragments. Conditioned medium <strong>of</strong> four replicates<br />

<strong>of</strong> non-stimulated, metabolically inactivated (MI) or cytokines stimulated explants from day 7 or day 21 were separately pooled by days and either<br />

left untreated (A and C) or deglycosylated (B and D). Pooled supernatants were run in 3–8% precasted Tris–acetate gels under reducing conditions.<br />

The two uniform membranes were stained with either the monoclonal antibody BC-3 (1:50 dilution) staining for the aggrecanase-mediated<br />

374 ARGSVI neoepitope (A and B) or the monoclonal antibody AF-28 (1200 ng ml −1 ), staining for the MMP-mediated 342 FFGVG-sequence (C and D).<br />

Peroxidase-labelled rabbit antimouse antibody (1:5000 dilution) was used as a secondary antibody. A standard molecular weight marker was also<br />

run to determine the size <strong>of</strong> the detected fragments (not shown). W/O, non-stimulated controls).<br />

nM Aggrecanase Conc.<br />

/mg cartilage<br />

0.7<br />

0.6<br />

0.5<br />

0.4<br />

0.3<br />

0.2<br />

0.1<br />

0.0<br />

W/O<br />

MI<br />

Cytokines<br />

Day 7<br />

**<br />

Day 21<br />

Figure 4. Quantification <strong>of</strong> aggrecanase activity in catabolically<br />

stimulated bovine explants. Bovine articular cartilage explants were<br />

isolated and cultured in the presence or absence <strong>of</strong> cytokines for 21<br />

days. Non-stimulated explants were used as the vehicle. Metabolically<br />

inactivated (MI) explants were used as a negative control to measure<br />

the non-chondrocyte-mediated release. The mean value <strong>of</strong> aggrecanase<br />

activity in the conditioned medium was determined by measuring<br />

four different wells containing the same treatment. This was done<br />

for all three different treatments at days 7 and 21. The aggrecanase<br />

activity was significant increased (p < 0.01) in supernatants (n = 4) from<br />

cytokine-stimulated cartilage compared with the non-stimulated controls<br />

(W/O) at day 21.<br />

83<br />

The conditioned medium from cytokine-stimulated<br />

explants from day 21 was digested in vitro with<br />

ADAMTS-4 and measured by the 342 FFGVG-G2 and<br />

374 ARGSV-G2 assays. The aggrecanase degradation index<br />

( 374 ARGSV-G2/ 342 FFGVG-G2) was calculated and the<br />

ADAMTS-4 digested media produced a 225% elevation<br />

compared with the index from non-ADAMTS-4-digested<br />

media (Figure 5). This indicates that the 373/374 cleavage<br />

site in the aggrecan fragments present at day 21 (only<br />

low-molecular-weight aggrecan) was functional, but only<br />

cleaved in vitro, not ex vivo.<br />

Substrate affinities by MMPs and aggrecanases may<br />

be essential for the diverse generation <strong>of</strong> 342 FFGVG-G2<br />

and 374 ARGSV-G2, and not the availability <strong>of</strong> aggrecan<br />

But why were the aggrecanase-mediated aggrecan<br />

fragments not present ex vivo at day 21, when both<br />

aggrecanases and a functional aggrecanase cleavage site<br />

were present in the conditioned medium? To investigate<br />

the difference <strong>between</strong> in vitro and ex vivo cleavage in<br />

aggrecan, we focused on the importance <strong>of</strong> glycosylation,<br />

which was absent in the late culture period. We investigated<br />

the different proteolytic potentials for ADAMTS-4<br />

and MMP-9 on either native or deglycosylated aggrecan<br />

in vitro and compared the results with the present ex vivo<br />

study. These proteolytic digestions were assessed by the<br />

374 ARGSV-G2 and 342 FFGVG-G2 assays.


Biomarkers Downloaded from informahealthcare.com by Panum Biblioteket on 08/31/10<br />

For personal use only.<br />

272 Suzi Hoegh Madsen et al.<br />

ADAMTS-4 digestion significantly increased the levels<br />

<strong>of</strong> 374 ARGSV-G2 on both native and deglycosylated<br />

aggrecan compared with the non-digested controls<br />

(Figure 6A). Furthermore, the 374 ARGSV-G2 levels were<br />

( 374 ARGSVI-G2)/( 342 FFGVG-G2)<br />

1.2<br />

1.0<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0.0<br />

Cytokines<br />

Day 21<br />

*<br />

Cytokines +<br />

ADAMTS-4<br />

Figure 5. Further cleavage <strong>of</strong> released matrix metalloproteinase<br />

(MMP)-mediated 342 FFGVG-G2 fragments to 374 ARGSVI-G2 by<br />

ADAMTS-4. Supernatants from bovine articular cartilage explants cultured<br />

in the presence <strong>of</strong> cytokines for 21 days (n = 4) were digested with<br />

20 nM ADAMTS-4, diluted 1:50 and assessed by the immunoassays<br />

374 ARGSV-G2 and 342 FFGVG-G2 quantifying aggrecanase- and MMPmediated<br />

aggrecan fragments, respectively. The degradation index<br />

( 374 ARGSV-G2)/( 342 FFGVG-G2) was made for the ADAMTS-4-treated<br />

cytokine-stimulated supernatants (black bar) and the undigested<br />

(ADAMTS) cytokine-stimulated control (grey bar); *p < 0.05.<br />

A<br />

ng 374-G2/ml<br />

250000<br />

200000<br />

150000<br />

100000<br />

50000<br />

0<br />

Nativ, ADAMSTS −<br />

ADAMTS mediated<br />

aggrecan degradation<br />

***<br />

Nativ, ADAMSTS +<br />

***<br />

Deglyco, ADAMSTS −<br />

***<br />

Deglyco, ADAMSTS +<br />

significantly higher for digested deglycosylated aggrecan<br />

compared with digested native aggrecan. MMP-9<br />

digestion showed a similar degradation pattern to<br />

ADAMTS-4 digestion. The levels <strong>of</strong> 342 FFGVG-G2 were<br />

significantly increased on both native and deglycosylated<br />

aggrecan compared with the non-digested controls<br />

(Figure 6B). Furthermore, the 342 FFGVG-G2 levels<br />

were significantly higher for digested deglycosylated<br />

aggrecan compared with digested native aggrecan.<br />

This did not correlate with the detection <strong>of</strong> 374 ARGSV-G2<br />

fragments at day 7 (high level) and day 21 (low level)<br />

in the ex vivo study (Figure 2A), but highlights the<br />

importance <strong>of</strong> distinguishing <strong>between</strong> in vitro and ex<br />

vivo results.<br />

Interestingly, the 374 ARGSV-G2 levels were approximately<br />

50 times greater than the 342 FFGVG-G2 levels.<br />

This detectable difference <strong>between</strong> the two neoepitope<br />

assays measured from in vitro samples resembles the<br />

difference we were able to detect in the individual peaks<br />

at days 7 and 21 in the ex vivo samples (Figure 2A and B).<br />

This indicates that the ex vivo measurements were reliable.<br />

This confirms that the lower concentration level <strong>of</strong><br />

342 FFGVG-G2 compared with 374 ARGSV-G2 was not due<br />

to limited aggrecan as a substrate.<br />

We were unable to conclude if glycosylation played<br />

an <strong>important</strong> role, based on the in vitro study. However,<br />

the substrate affinities <strong>of</strong> MMPs and aggrecanases may<br />

still be the essential factor for the diverse generation <strong>of</strong><br />

342 FFGVG-G2 and 374 ARGSV-G2 ex vivo.<br />

B<br />

ng 342-G2/ml<br />

4000<br />

3000<br />

2000<br />

1000<br />

0<br />

Nativ, MMP −<br />

MMP mediated<br />

aggrecan degradation<br />

Figure 6. Matrix metalloproteinase (MMP)-9 and ADAMTS-4 digestion on native and deglycosylated aggrecan. Purified bovine aggrecan was<br />

either left untreated or deglycosylated. The native and deglycosylated aggrecan were each digested with MMP-9 and ADAMTS-4 and assessed by<br />

the 374 ARGSV-G2 (A) and 342 FFGVG-G2 (B) ELISA assays; *p < 0.001; n = 4.<br />

84<br />

***<br />

Nativ, MMP +<br />

***<br />

Deglyco, MMP −<br />

***<br />

Deglyco, MMP +


Biomarkers Downloaded from informahealthcare.com by Panum Biblioteket on 08/31/10<br />

For personal use only.<br />

Discussion<br />

To elucidate the molecular mechanisms behind the<br />

destruction <strong>of</strong> the ECM, a large number <strong>of</strong> studies have<br />

reported the molecular nature <strong>of</strong> type II collagen and<br />

aggrecan fragments as they are released into the synovial<br />

fluid (Malfait et al. 2002, Sandy et al. 1992, Struglics<br />

et al. 2006) or into explant culture supernatants (Caterson<br />

et al. 2000, Fosang et al. 2000, Little et al. 1999, Sumer<br />

et al. 2007, Sztrolovics et al. 2002). While members <strong>of</strong> the<br />

MMP family have long been known to have the capacity<br />

to cleave aggrecan (Fosang et al. 1996, Struglics et al.<br />

2006), only recently were the aggrecanases identified to<br />

have the same ability (Stanton et al. 2005, Struglics et al.<br />

2006). Subsequently, the relative importance <strong>of</strong> MMPs<br />

and aggrecanases in aggrecanolysis has been debated.<br />

In the present study we demonstrated that in cytokinestimulated<br />

ex vivo cultures <strong>of</strong> articular bovine cartilage,<br />

both MMP- and aggrecanase-mediated aggrecan fragments<br />

were released into the supernatant, but not<br />

simultaneously. The catabolic stimulation <strong>of</strong> cartilage<br />

explants induced an early release <strong>of</strong> aggrecanase-mediated<br />

374 ARGSVI-G2 fragments, and only after returning to<br />

background levels did we detect an increase in the MMPmediated<br />

release <strong>of</strong> 342 FFGVG-G2 fragments. The peak at<br />

day 7 and subsequent drop in the release <strong>of</strong> aggrecanasemediated<br />

aggrecan fragments in the ex vivo culture<br />

(Figure 2A) was not caused by depletion <strong>of</strong> the aggrecan<br />

as a substrate, because we detected MMP-mediated<br />

aggrecan degradation later in the culture period (Figure<br />

2B), which verified the presence <strong>of</strong> the core proteins <strong>of</strong><br />

aggrecan. Furthermore, our data suggested that the drop<br />

in aggrecanase-mediated aggrecan fragments was not<br />

caused by the lack <strong>of</strong> the aggrecanases, as a high level<br />

<strong>of</strong> their activity was detected in the late culture period<br />

(Figure 4).<br />

As presented, we found both evidence <strong>of</strong> the presence<br />

<strong>of</strong> aggrecan as a substrate and aggrecanase activity in the<br />

late period <strong>of</strong> culture, but interestingly no 374 ARGSVI-G2<br />

fragments. We further demonstrated that it was possible<br />

to generate 374 ARGSVI-G2 fragments from the MMPmediated<br />

342 FFGVG-G2 fragments, when treated with<br />

aggrecanases in vitro. This suggests that low concentrations<br />

<strong>of</strong> 374 ARGSVI-G2 fragments in the late culture period<br />

did not result from an inaccessible 373/374-cleavage site<br />

in the 342 FFGVG-G2 fragments. However, this does not<br />

explain why the cleavage only happens in vitro and not<br />

ex vivo. The ambiguity regarding the lack <strong>of</strong> 374 ARGSVI-G2<br />

fragments in the late culture period resulted in further<br />

investigations. We detected an identical pattern <strong>of</strong><br />

aggrecanase-mediated aggrecan degradation as occurred<br />

with the sGAG release. This link <strong>between</strong> 374 ARGSVI-G2<br />

fragments and sGAGs was confirmed via Western blotting,<br />

showing highly glycosylated 374 ARGSVI-G2 fragments<br />

from the early culture period and deglycosylated<br />

Aggrecanase- and MMP-mediated aggrecan degradation 273<br />

85<br />

342 FFGVG-G2 fragments in the late culture period. These<br />

data suggest that two different pools <strong>of</strong> aggrecan molecules<br />

contain an N-terminal neoepitope in the IGD<br />

region ( 342 FFGVG and 374 ARGSVI), the G2 domain and a<br />

C-terminus <strong>of</strong> considerable difference. In particular, the<br />

C-terminus and glycosylation are obvious candidates to<br />

explain the discrepancy in the molecular sizes <strong>of</strong> the two<br />

aggrecan pools. The results from the present paper are<br />

summarized in Figure 7.<br />

Our high- versus low-molecular-weight data support<br />

the theory described by Sandy (2006) that MMPs<br />

and aggrecanases act independently, and aggrecanases<br />

cleave intact aggrecan, whereas MMPs degrade aggrecan<br />

molecules which have already been C-terminally cleaved.<br />

Moreover, Lark et al. (1997) reported that MMPs are<br />

located in the damaged cartilage area, whereas aggrecanases<br />

are located in the non-damaged area. This is<br />

consistent with our study showing that aggrecanases are<br />

present in the beginning <strong>of</strong> the culture period and MMPs<br />

are present in the later culture period when the cartilage<br />

is further degraded.<br />

Struglics et al. (2006) suggested that two distinct pathways<br />

were responsible for aggrecanolysis in vivo: first,<br />

the MMP-mediated 342 FFGVG-neoepitope in the pericellular<br />

matrix and secondly the aggrecanase-mediated<br />

374 ARGSVI neoepitope in the inter-territorial matrix, on<br />

the same aggrecan molecule. This is not supported by<br />

the results in the present paper. However, the theory <strong>of</strong><br />

the two distinct pathways is consistent with observations<br />

made in the present study, and is furthermore supported<br />

by identification <strong>of</strong> such aggrecan fragments in human<br />

synovial fluid spanning molecular sizes <strong>of</strong> 80–335 kDa<br />

(Struglics et al. 2006). Our Western blot data differ from<br />

those <strong>of</strong> Struglics et al. (2006) who found MMP-mediated<br />

high-molecular-weight fragments in the synovial fluid<br />

samples and in cartilage explants stimulated with MMPs.<br />

We only detected high-molecular-weight fragments from<br />

aggrecanase-mediated degradation (Figure 3).<br />

One possible explanation for this interesting discrepancy<br />

is the differences <strong>between</strong> ex vivo and in vivo settings.<br />

In the current experiments we used a synchronic<br />

culture system, which presents a temporal predefined<br />

order <strong>of</strong> events following induction <strong>of</strong> catabolic activity<br />

against bovine cartilage (Karsdal 2007, 2008). This<br />

experimental setting is in contrast to OA patients who<br />

may have cartilage mal-metabolism ranging in severity at<br />

different distinct local places in the same joint. Enzymes<br />

are to some extent freely diffusible in the synovial fluid,<br />

so damaged cartilage areas mediated by extensive MMP<br />

activity may also ‘share’ some <strong>of</strong> these MMPs with other<br />

areas which have less cartilage damage, and which may<br />

primarily consist <strong>of</strong> aggrecanase activity. Such ‘enzyme<br />

sharing’ in the local environment may indeed generate<br />

the MMP-mediated high-molecular-weight fragments as<br />

described by Struglics et al. (2006).


Biomarkers Downloaded from informahealthcare.com by Panum Biblioteket on 08/31/10<br />

For personal use only.<br />

274 Suzi Hoegh Madsen et al.<br />

Degradation fragments or activity<br />

Time<br />

Importantly, Struglics et al. (2006) also suggested that<br />

aggrecanases cleaved MMP-mediated fragments in the<br />

highly glycosylated region (<strong>between</strong> G2 and G3), and not<br />

in the IGD. In addition, they presented data in agreement<br />

with Fosang et al. (2000), who clearly demonstrated that<br />

aggrecanases were unable to cleave 342 FFGVG fragments<br />

at the NITEGE 373–374 ARGSVI site (Fosang et al. 2000). Our<br />

data support these observations and conclusions ex vivo,<br />

albeit not in vitro, as we observed that ADAMTS-4 was<br />

capable <strong>of</strong> processing the MMP-mediated 342 FFGVG-G2<br />

fragments further in the IGD (Figure 5). However, a high<br />

concentration <strong>of</strong> aggrecanases was used to post-cleave<br />

the fragments released into the supernatant, and thus<br />

our data suggest that these fragments released ex vivo<br />

were protected from aggrecanase-mediated cleavage at<br />

the NITEGE 373–374 ARGSVI site. The molecular mechanism<br />

behind this protection ex vivo is currently debated, but<br />

it may be the lack <strong>of</strong> glycosylation <strong>of</strong> the MMP-mediated<br />

aggrecan fragments. We support the suggestion by<br />

Tottorella et al. (2000) that aggrecanases may favour<br />

highly glycosylated forms <strong>of</strong> aggrecan, or depend on<br />

the protein sequence upstream <strong>of</strong> the 374 ARGSVI site<br />

for proper recognition and processing ex vivo. In fact,<br />

another group reported that aggrecanase activity is promoted<br />

by the presence <strong>of</strong> glycosylated aggrecan (Pratta<br />

et al. 2000).<br />

MMP activity<br />

Aggrecanase<br />

activity<br />

342-G2<br />

sGAG<br />

374-G2<br />

Figure 7. Schematic overview <strong>of</strong> aggrecanolysis in the explants model. The dotted lines represent protease activity <strong>of</strong> matrix metalloproteinase<br />

(MMPs) and aggrecanases over time. The other lines represent the degradation fragments released from the cartilage explants to the conditioned<br />

medium over time. Above the lines are cartoons <strong>of</strong> aggrecan attached to hyaluronic acid, which show the degradation and release <strong>of</strong> sulfated glycosaminoglycans<br />

(sGAGs) from the aggrecan molecule over time. Aggrecanases cleave the glycosylated aggrecan molecules in the early culture<br />

period, whereas MMPs degrade deglycosylated aggrecan molecules in the late culture period.<br />

86<br />

All the aforementioned biomarkers were measured in<br />

conditioned medium synthesized by ex vivo cleavage. We<br />

designed an in vitro experiment to investigate the differences<br />

<strong>of</strong> in vitro and ex vivo cleavage in aggrecan. This<br />

enabled us to investigate the different proteolytic affinities<br />

for ADAMTS-4 and MMP-9 on native or deglycosylated<br />

aggrecan, which we assessed by the 374 ARGSV-G2 and<br />

342 FFGVG-G2 assays. The measurements showed significant<br />

increased levels for 374 ARGSV-G2 and 342 FFGVG-G2<br />

fragments when stimulated with ADAMTS-4 and MMP-9,<br />

respectively. In vitro cleavage showed that 374 ARGSV-G2<br />

levels were approximately 50 times greater than the<br />

342 FFGVG-G2 levels. This difference <strong>between</strong> the two<br />

neoepitope assays measured in the in vitro samples<br />

resembled the difference we detected in ex vivo samples<br />

(Figure 2A and B). This suggests that the approximately<br />

ten times lower levels <strong>of</strong> 342 FFGVG-G2 compared with<br />

374 ARGSV-G2 levels measured from the ex vivo cleavage,<br />

were not due to limited aggrecan as a substrate, and therefore<br />

the measurements ex vivo represent a reproducible<br />

difference <strong>between</strong> the neoepitopes. Interestingly, the in<br />

vitro measurements showed that ADAMTS-4 and MMP-9<br />

both show a preference to degrade deglycosylated aggrecan<br />

rather than native aggrecan. This is somewhat different<br />

from the ex vivo measurements, which indicated<br />

that ADAMTS-4 prefer to degrade glycosylated aggrecan


Biomarkers Downloaded from informahealthcare.com by Panum Biblioteket on 08/31/10<br />

For personal use only.<br />

and MMP-9 favoured degrading deglycosylated aggrecan<br />

(Figures 2 and 3). This highlights the need to distinguish<br />

<strong>between</strong> in vitro and ex vivo results.<br />

In conclusion, this study highlights the importance <strong>of</strong><br />

the separate and distinct proteolytic processing <strong>of</strong> aggrecan,<br />

which varies over time. If the current findings are<br />

to be useful in a clinical setting, it may be favourable to<br />

target activity or expression <strong>of</strong> aggrecanases early in the<br />

OA process, whereas at later stages, after the depletion <strong>of</strong><br />

sGAGs, it may be more efficacious to target expression or<br />

activity <strong>of</strong> MMPs.<br />

Acknowledgements<br />

This study was supported in part by The Ministry <strong>of</strong><br />

Science Technology and Innovation, Denmark.<br />

Declaration <strong>of</strong> interest<br />

All authors declare that the affiliation declares full disclosure.<br />

In addition, Drs Karsdal and Qvist own stock in<br />

Nordic Bioscience.<br />

References<br />

Behrens F, Kraft EL, Oegema TR Jr. (1989). Biochemical changes in<br />

articular cartilage after joint immobilization by casting or external<br />

fixation. J Orthop Res 7:335–43.<br />

Caterson B, Flannery CR, Hughes CE, Little CB. (2000). Mechanisms<br />

involved in cartilage proteoglycan catabolism Matrix Biol<br />

19:333–44.<br />

Felson DT, Neogi T. (2004). Osteoarthritis: is it a disease <strong>of</strong> cartilage or<br />

<strong>of</strong> <strong>bone</strong>? Arthritis Rheum 50:341–4.<br />

Flannery CR, Lark MW, Sandy JD. (1992). <strong>Identification</strong> <strong>of</strong> a stromelysin<br />

cleavage site within the interglobular domain <strong>of</strong> human<br />

aggrecan. Evidence for proteolysis at this site in vivo in human<br />

articular cartilage. J Biol Chem 267:1008–14.<br />

Fosang AJ, Last K, Gardiner P, Jackson DC, Brown L. (1995).<br />

Development <strong>of</strong> a cleavage-site-specific monoclonal antibody<br />

for detecting metalloproteinase-derived aggrecan fragments:<br />

detection <strong>of</strong> fragments in human synovial fluids. Biochem J<br />

310:337–43.<br />

Fosang AJ, Last K, Maciewicz RA. (1996). Aggrecan is degraded by<br />

matrix metalloproteinases in human arthritis. Evidence that<br />

matrix metalloproteinase and aggrecanase activities can be independent.<br />

J Clin Invest 98:2292–9.<br />

Fosang AJ, Last K, Stanton H, Weeks DB, Campbell IK, Hardingham<br />

TE, Hembry RM. (2000). Generation and novel distribution <strong>of</strong><br />

matrix metalloproteinase-derived aggrecan fragments in porcine<br />

cartilage explants. J Biol Chem 275:33027–37.<br />

Glasson SS, Askew R, Sheppard B, Carito B, Blanchet T, Ma HL,<br />

Flannery CR, Peluso D, Kanki K, Yang Z, Majumdar MK,<br />

Morris EA. (2005). Deletion <strong>of</strong> active ADAMTS5 prevents cartilage<br />

degradation in a murine model <strong>of</strong> osteoarthritis. Nature<br />

434:644–8.<br />

Hardingham TE, Fosang AJ. (1995). The structure <strong>of</strong> aggrecan and its<br />

turnover in cartilage. J Rheumatol Suppl 43:86–90.<br />

Ilic MZ, Haynes SR, Winter GM, Handley CJ. (1995). Kinetics <strong>of</strong> release<br />

<strong>of</strong> aggrecan from explant cultures <strong>of</strong> bovine cartilage from different<br />

sources and from animals <strong>of</strong> different ages. Acta Orthop<br />

Scand Suppl 266:33–7.<br />

Aggrecanase- and MMP-mediated aggrecan degradation 275<br />

87<br />

Karsdal MA, Madsen SH, Christiansen C, Henriksen K, Fosang AJ,<br />

Sondergaard BC. (2008). Cartilage degradation is fully reversible<br />

in the presence <strong>of</strong> aggrecanase but not matrix metalloproteinase<br />

activity. Arthritis Res Ther 10:R63.<br />

Karsdal MA, Sumer EU, Wulf H, Madsen SH, Christiansen C, Fosang<br />

AJ, Sondergaard BC. (2007). Induction <strong>of</strong> increased cAMP levels<br />

in articular chondrocytes blocks matrix metalloproteinasemediated<br />

cartilage degradation but not aggrecanase-mediated<br />

cartilage degradation. Arthritis Rheum 56:1549–58.<br />

Kiani C, Chen L, Wu YJ, Yee AJ, Yang BB. (2002). Structure and function<br />

<strong>of</strong> aggrecan. Cell Res 12:19–32.<br />

Lark MW, Bayne EK, Flanagan J, Harper CF, Hoerrner LA, Hutchinson<br />

NI, Singer II, Donatelli SA, Weidner JR, Williams HR, Mumford<br />

RA, Lohmander LS. (1997). Aggrecan degradation in human cartilage.<br />

Evidence for both matrix metalloproteinase and aggrecanase<br />

activity in normal osteoarthritic and rheumatoid joints. J<br />

Clin Invest 100:93–106.<br />

Little CB, Flannery C R, Hughes CE, Mort JS, Roughley PJ, Dent C,<br />

Caterson B. (1999). Aggrecanase versus matrix metalloproteinases<br />

in the catabolism <strong>of</strong> the interglobular domain <strong>of</strong> aggrecan<br />

in vitro. Biochem J 344:61–8.<br />

Lohmander S, Antonopoulos CA, Friberg U. (1973). Chemical and<br />

metabolic heterogeneity <strong>of</strong> chondroitin sulfate and keratin<br />

sulfate in guinea pig cartilage and nucleus pulposus. Biochim<br />

Biophys Acta 304:430–48.<br />

Maehara H, Suzuki K, Sasaki T, Oshita H, Wada E, Inoue T, Shimizu<br />

K. (2007). G1-G2 aggrecan product that can be generated by<br />

M-calpain on truncation at Ala709-Ala710 is present abundantly<br />

in human articular cartilage. J Biochem 141:469–77.<br />

Malfait AM, Liu RQ, Ijiri K, Komiya S, Tortorella MD. (2002). Inhibition<br />

<strong>of</strong> ADAM-TS4 and ADAM-TS5 prevents aggrecan degradation in<br />

osteoarthritic cartilage. J Biol Chem 277:22201–8.<br />

Mercuri FA, Maciewicz RA, Tart J, Last K, Fosang AJ. (2000). Mutations<br />

in the interglobular domain <strong>of</strong> aggrecan alter matrix metalloproteinase<br />

and aggrecanase cleavage patterns. Evidence that matrix<br />

metalloproteinase cleavage interferes with aggrecanase activity.<br />

J Biol Chem 275:33038–45.<br />

Mok SS, Masuda K, Hauselmann HJ, Aydelotte MB, Thonar EJ. (1994).<br />

Aggrecan synthesized by mature bovine chondrocytes suspended<br />

in alginate. <strong>Identification</strong> <strong>of</strong> two distinct metabolic matrix pools.<br />

J Biol Chem 269:33021–7.<br />

Oshita H, Sandy JD, Suzuki K, Akaike A, Bai Y, Sasaki T, Shimizu K.<br />

(2004). Mature bovine articular cartilage contains abundant<br />

aggrecan that is C-terminally truncated at Ala719-Ala720 a site<br />

which is readily cleaved by m-calpain. Biochem J 382:253–9.<br />

Pratta MA, Tortorella MD, Arner EC. (2000). Age-related changes in<br />

aggrecan glycosylation affect cleavage by aggrecanase. J Biol<br />

Chem 275:39096–102.<br />

Roughley PJ, Lee ER. (1994). Cartilage proteoglycans: structure and<br />

potential functions. Microsc Res Tech 28:385–97.<br />

Rousseau JC, Sumer EU, Hein G, Sondergaard BC, Madsen SH,<br />

Pedersen C, Neumann T, Mueller A, Qvist P, Delmas P, Karsdal<br />

MA. (2008). Patients with rheumatoid arthritis have an altered<br />

circulatory aggrecan pr<strong>of</strong>ile. BMC Musculoskelet Disord 9:74.<br />

Sandy JD. (2006). A contentious issue finds some clarity: on the<br />

independent and complementary roles <strong>of</strong> aggrecanase activity<br />

and MMP activity in human joint aggrecanolysis. Osteoarthritis<br />

Cartilage 14:95–100.<br />

Sandy JD, Flannery CR, Neame PJ, Lohmander LS. (1992). The structure<br />

<strong>of</strong> aggrecan fragments in human synovial fluid. Evidence<br />

for the involvement in osteoarthritis <strong>of</strong> a novel proteinase which<br />

cleaves the Glu 373-Ala 374 bond <strong>of</strong> the interglobular domain. J<br />

Clin Invest 89:1512–16.<br />

Sondergaard BC, Henriksen K, Wulf H, Oestergaard S, Schurigt U,<br />

Brauer R, Danielsen I, Christiansen C, Qvist P, Karsdal MA.<br />

(2006a). Relative contribution <strong>of</strong> matrix metalloproteinase and<br />

cysteine protease activities to cytokine-stimulated articular cartilage<br />

degradation. Osteoarthritis Cartilage 14:738–48.<br />

Sondergaard BC, Wulf H, Henriksen K, Schaller S, Oestergaard S, Qvist<br />

P, Tanko LB, Bagger YZ, Christiansen C, Karsdal MA. (2006b).<br />

Calcitonin directly attenuates collagen type II degradation by<br />

inhibition <strong>of</strong> matrix metalloproteinase expression and activity<br />

in articular chondrocytes. Osteoarthritis Cartilage 14:759–68.


Biomarkers Downloaded from informahealthcare.com by Panum Biblioteket on 08/31/10<br />

For personal use only.<br />

276 Suzi Hoegh Madsen et al.<br />

Stanton H, Rogerson FM, East CJ, Golub SB, Lawlor KE, Meeker CT,<br />

Little CB, Last K, Farmer PJ, Campbell IK, Fourie AM, Fosang AJ.<br />

(2005). ADAMTS5 is the major aggrecanase in mouse cartilage in<br />

vivo and in vitro. Nature 434:648–52.<br />

Struglics A, Larsson S, Pratta MA, Kumar S, Lark MW, Lohmander<br />

LS. (2006). Human osteoarthritis synovial fluid and joint cartilage<br />

contain both aggrecanase- and matrix metalloproteinase-generated<br />

aggrecan fragments. Osteoarthritis Cartilage<br />

14:101–13.<br />

Sumer EU, Sondergaard BC, Rousseau JC, Delmas PD, Fosang AJ,<br />

Karsdal MA, Christiansen C, Qvist P. (2007). MMP and non-<br />

MMP-mediated release <strong>of</strong> aggrecan and its fragments from<br />

articular cartilage: a comparative study <strong>of</strong> three different<br />

88<br />

aggrecan and glycosaminoglycan assays. Osteoarthritis Cartilage<br />

15:212–21.<br />

Sztrolovics R, White RJ, Roughley PJ, Mort JS. (2002). The mechanism<br />

<strong>of</strong> aggrecan release from cartilage differs with tissue origin and<br />

the agent used to stimulate catabolism. Biochem J 362:465–72.<br />

Tortorella M, Pratta M, Liu RQ, Abbaszade I, Ross H, Burn T, Arner E.<br />

(2000). The thrombospondin motif <strong>of</strong> aggrecanase-1 (ADAMTS-4)<br />

is critical for aggrecan substrate recognition and cleavage. J Biol<br />

Chem 275:25791–7.<br />

Tortorella MD, Burn TC, Pratta MA, Abbaszade I, Hollis JM, Liu R,<br />

Rosenfeld SA, Copeland RA, Decicco CP, Wynn R and others.<br />

(1999). Purification and cloning <strong>of</strong> aggrecanase-1: a member <strong>of</strong><br />

the ADAMTS family <strong>of</strong> proteins. Science 284:1664–6.


CHAPTER 7<br />

CHAPTER 7: PAPER IV<br />

PAPER IV<br />

___________________________________________________________________________<br />

Introductory remarks<br />

This manuscript describes the endogenous proteases in human OA cartilage, and the biochemical<br />

markers that are able to assess the different proteolytic pathways for both collagen type II and<br />

aggrecan degradation. More investigation on this topic may have significant value for<br />

development <strong>of</strong> new treatments for OA.<br />

89


CHAPTER 7: PAPER IV<br />

Metalloproteinase and cysteine protease<br />

pr<strong>of</strong>iling in normal and Osteoarthritic cartilage<br />

Suzi Hoegh Madsen 1 , Kim Henriksen 3 , Søren Tvorup Christensen 2 , Morten Asser Karsdal 1 , and<br />

Anne-Christine Bay-Jensen 1<br />

In review in Proteome Science<br />

1 Cartilage Biology and Biomarkers R&D, Nordic Bioscience A/S, Herlev, Denmark<br />

2 Department <strong>of</strong> Molecular Biology, University <strong>of</strong> Copenhagen, Copenhagen, Denmark<br />

3 Bone Biology R&D, Nordic Bioscience A/S, Herlev, Denmark<br />

___________________________________________________________________________<br />

Abstract<br />

Treatment for cartilage degenerating diseases is either analgesic or joint replacement. Thus, the<br />

discovery and development <strong>of</strong> new treatments are needed. Proteases, such as metalloproteinases<br />

and cathepsins, play a major role in the net degradation <strong>of</strong> cartilage, and are therefore a natural<br />

target for drug development. By testing the ability <strong>of</strong> a library <strong>of</strong> proteases to degrade cartilage<br />

explants, and by inhibiting one type <strong>of</strong> protease, we aim to find the proteases responsible for<br />

collagen type II and aggrecan degradation and possible masking effects by other proteases.<br />

The cysteine inhibitor, E64, and the proteases; MMP-3 and -7, and ADAMTS-4<br />

and -5, all increased aggrecan degradation (AGNx-II), in human metabolic inactivated<br />

osteoarthritic (OA) cartilage. The MMP inhibitor, GM6001, abrogated all the increased aggrecan<br />

degradation. Collagen type II was degraded with MMP-7 (CTX-II) and MMP-9 (NBC2), which<br />

both could be inhibited by GM6001. Human OA cartilage cultured in buffer optimal for cysteine<br />

proteases showed an increase in levels <strong>of</strong> CTX-II and NBC2 fragments when incubated with<br />

GM6001. Furthermore, we observed altered levels <strong>of</strong> the biomarkers <strong>between</strong> normal and pre-<br />

cytokine-stimulated bovine cartilage, indicating an induced alteration <strong>of</strong> the endogenous<br />

proteases.<br />

In human OA cartilage, the degradation markers are released in part by different<br />

proteolytic pathways. Inhibition <strong>of</strong> cysteine proteases unmasks the levels <strong>of</strong> MMP-mediated<br />

aggrecan degradation in OA cartilage. Inhibition <strong>of</strong> MMPs also unmasks the levels <strong>of</strong> the collagen<br />

type II degradation mediated by non-MMPs. The role <strong>of</strong> endogenous proteases probably<br />

depends on the stage <strong>of</strong> OA, as we did not observe the same release <strong>of</strong> biomarkers in normal<br />

versus cytokine-stimulated and OA cartilage.<br />

90


Introduction<br />

CHAPTER 7: PAPER IV<br />

Cartilage turnover is a complex process in which proteases play a prominent role in both health<br />

and disease. The extracellular matrix (ECM) in healthy articular cartilage is expressed and<br />

regulated by the chondrocytes, which include expression <strong>of</strong> matrix proteins, proteases, and<br />

protease inhibitors 1 . One <strong>of</strong> the major mediators <strong>of</strong> cartilage destruction in osteoarthritis (OA) is<br />

the metalloproteinases; matrix metalloproteinases (MMPs) and A Disintegrin And<br />

Metalloproteinase with Thrombospondin Motifs (ADAMTS’) 2 . In few words, the destruction <strong>of</strong><br />

ECM in OA results from initial aggrecan degradation by ADAMTS’ and subsequently<br />

degradation by diverse MMPs that continues to degrade the major components <strong>of</strong> the ECM, e.g.<br />

collagen type II and aggrecan 2,3,4 . However, cysteine proteases are also essential players in the<br />

proteolytic cleavage <strong>of</strong> ECM in OA, which are synthesized both by the chondrocytes and<br />

synovial cells in response to cytokines and growth factors 5 .<br />

Currently, treatment <strong>of</strong> cartilage degenerative diseases is either analgesic or joint<br />

replacement. Thus, the discovery and development <strong>of</strong> new treatments are needed. Since proteases<br />

- such as MMPs and cathepsins - play a major role in the net degradation <strong>of</strong> cartilage, they are a<br />

natural target for drug development. Early treatment with protease inhibitors to either reduce<br />

inflammation and/or reduce aggrecan/collagen loss may have the potential to reduce the onset<br />

<strong>of</strong> future idiopathic or posttraumatic osteoarthritis. However, to target the right protease at the<br />

right time during the progression <strong>of</strong> OA, a better understanding <strong>of</strong> the proteolytic processes is<br />

needed. ECM-neoepitope biomarkers are great tools for investigating the proteolytic processes 6 .<br />

The collagen type II degradation marker, C-terminal telopeptide degradation fragment (CTX-II) 7<br />

and the aggrecan degradation marker, AGNx-II 8 , are both MMP-specific. The novel collagen<br />

type II intra-helical degradation markers, NBC2, are supposed to represent cathepsin-specificity.<br />

However, the biomarker will be evaluated in the present study.<br />

In this study, we identified which MMPs, ADAMTS’ and cathepsins were able to<br />

generate CTX-II, NBC2, and AGNx-II fragments using an in vitro and ex vivo model <strong>of</strong> human<br />

OA cartilage. Such data could provide information on the specificity <strong>of</strong> the different proteases<br />

for cleavage <strong>of</strong> collagen and aggrecan, and also inform us about the proteolytic pathways for each<br />

biomarker release. Furthermore, the endogenous proteases in normal bovine cartilage were<br />

compared with cytokine-stimulated bovine and human OA cartilage. The cytokines, oncostatin M<br />

(OSM) and tumor necrosis factor-α (TNF-α), are inducers <strong>of</strong> cartilage degradation in vivo and in<br />

vitro 9 , and would allow us to induce a controlled catabolic system towards the one seen in human<br />

OA cartilage. I.e. induction <strong>of</strong> aggrecanases after 4 days with cytokine stimulation and induction<br />

<strong>of</strong> MMPs after 11 days with cytokine stimulation 3 . By inhibiting one type <strong>of</strong> protease, we aimed<br />

to find the proteases responsible for collagen type II and aggrecan degradation and possible<br />

masking effects by other proteases.<br />

91


Methods<br />

Sources <strong>of</strong> proteases, inhibitors and cytokines<br />

CHAPTER 7: PAPER IV<br />

Purified human matrix metalloproteinase-1 (MMP-1) and recombinant human MMP-7 were<br />

purchased from Enzo life science, DK. Recombinant human Cathepsin-L and purified human<br />

Cathepsin-S and Cathepsin-B were from BioVision, US. Recombinant human aggrecanases;<br />

ADAMTS-4 and ADAMTS-5, were from Chemicon international, DK. Purified human MMP-9<br />

and recombinant human MMP-3, MMP-13 and Cathepsin-K, were purchased from Biocol, DE.<br />

The matrix metalloprotease inhibitor (R)-N4-Hydroxy-N1-[(S)-2-(1H-indol-3-yl)-1-<br />

methylcarbamoyl-ethyl]-2-isobutyl-succinamide (GM6001) and the cysteine protease inhibitor<br />

trans-Epoxysuccinyl-L-leucylamido(4-guanidino)butane, L-trans-3-Carboxyoxiran-2-carbonyl-L-<br />

leucylagmatine, N-(trans-Epoxysuccinyl)-L-leucine 4-guanidinobutylamide (E64) were both<br />

purchased from Sigma, DK. The cytokines, oncostatin M (OSM) was from Sigma, DK, and the<br />

Tumor Necrosis Factor-α (TNF-α) was from R&D systems, UK.<br />

Buffers<br />

To evaluate endogenous activity <strong>of</strong> metalloproteinases (MMPs and ADAMTS’), cartilage explants<br />

were incubated in an MMP buffer [50 mM Tris-HCL, 200 mM NaCl, 5 mM CaCl 2, 1 µM ZnCl 2,<br />

0.05% NaN 3, 0.05% Brij-35, pH 7]. Activation <strong>of</strong> latent MMPs was performed by adding 1 mM<br />

freshly made p-Aminophenylmercurie (APMA). To evaluate endogenous activity <strong>of</strong> cysteine<br />

proteinases, cartilage explants were incubated in a cysteine buffer [100 mM Na-Acetate, 5 mM<br />

EDTA, 0.05% NaN 3, pH 5]. Activation <strong>of</strong> latent cysteine proteinases was performed by adding<br />

20 mM cysteine just before use. Both buffers were prepared with inspiration from Tabassi et al. 13 .<br />

Ex vivo models<br />

Human articular cartilage experiments<br />

The human OA cartilage was obtained from female patients <strong>between</strong> the ages <strong>of</strong> 65-77 years<br />

undergoing total knee arthroplasty. The patients were informed by both oral and written<br />

communication at the hospital and gave their written consent before entering study, which was<br />

approved by the Capital Region <strong>of</strong> Denmark, DK-3400, approval number HD-2007-0084. The<br />

cartilage was metabolically inactivated (MI) by freeze-thaw cycles and cut into homologous<br />

explants. Half <strong>of</strong> the MI explants were incubated in MMP buffer for 3, 6, and 9 h in the presence<br />

or absence <strong>of</strong> the inhibitors, GM6001 [10 µM] and E64 [40uM], and the proteases [100 ng/ml];<br />

MMP-1, -3, -7, -9, -13 and ADAMTS-4, -5. The other half were incubated in cysteine buffer for<br />

3, 6, and 9 h in the presence or absence <strong>of</strong> the inhibitors, GM6001 [10 µM] and E64 [40 µM], and<br />

the proteases [100 ng/ml]; Cathepsin-K, -B, -L, -S. At the end <strong>of</strong> each incubation time (3, 6, 9h),<br />

conditioned buffer (about 300 µl) was extracted and stored frozen at -20°C until assayed for<br />

specific biomarkers.<br />

92


Bovine articular cartilage experiments<br />

CHAPTER 7: PAPER IV<br />

Bovine knees (Hereford) were bought at the local slaughter house (Slangerup, DK) just after<br />

slaughtering. Articular cartilage explants were isolated under semi-sterile conditions and washed<br />

in PBS. One third <strong>of</strong> the cartilage explants were MI (t=0) and stored in -80°C until use. The rest<br />

<strong>of</strong> the cartilage explants were pre-cultured with pro-inflammatory cytokines, OSM [10 ng/ml]<br />

and TNF-α [20 ng/ml] (O+T), for 4 or 11 days to increase aggrecanase or MMP activity,<br />

respectively 3,4 . The cartilage explants pre-cultured in cytokines for 4 or 11 days were after each<br />

termination washed in PBS and MI, and stored in -80°C until use. The frozen explants without<br />

cytokine pre-culture (t=0) and the pre-cultured catabolic induced explants (t=4 and t=11), were<br />

thawed and incubated in MMP or cysteine buffer for 3, 6, and 9 h in the presence or absence <strong>of</strong><br />

the inhibitors, GM6001 [10 µM] and E64 [40 µM]. At the end <strong>of</strong> each incubation time (3h, 6h,<br />

9h), conditioned buffer (about 300 µl) was extracted and stored frozen at -20°C until assayed for<br />

specific biomarkers.<br />

Biomarkers <strong>of</strong> cartilage degradation<br />

The type II collagen telopeptide fragment, CTX-II (urine cartilaps, IDS Ltd, DK), were measured<br />

accordingly to the manufacturer’s instructions 7 . AGNx-II measures MMP-mediated aggrecan<br />

degradation accordingly to Sumer et al. 8 . The type II collagen inter helical neo-epitope fragment<br />

(…PKGARG 707 ), NBC2, was measured accordingly to Bay-Jensen et al. 2011 (unpublished).<br />

Statistic analysis<br />

Results in fig. 1 are shown as mean + standard error <strong>of</strong> mean (SEM). Statistic analysis in fig. 1<br />

was performed by ANOVA; nonparametric test, don’t assuming Gaussian distribution, n=4,<br />

Kruskal-Wallis test with Dunn’s multiple comparison post test. Statistic significance is indicated<br />

by the number <strong>of</strong> asterisks, * p < 0.05, ** p < 0.01, and *** p < 0.001. The Tables show mean<br />

value <strong>of</strong> n=4 ± standard deviation (SD).<br />

Results<br />

Detection <strong>of</strong> key proteases to degrade human OA cartilage ex vivo; evaluated by collagen type II<br />

degradation markers<br />

Human OA cartilage explants incubated in buffer optimal for MMPs, increased the levels <strong>of</strong><br />

CTX-II when incubated with MMP-7 (p


AGNx-II ng/mL<br />

CTX-II ng/mL<br />

NBC2 ng/mL<br />

2.4<br />

1.8<br />

1.2<br />

0.6<br />

Vehicle<br />

MMP1<br />

MMP3<br />

MMP buffer<br />

**<br />

MMP7<br />

MMP9<br />

MMP13<br />

CHAPTER 7: PAPER IV<br />

A B<br />

C<br />

E<br />

15<br />

10<br />

5<br />

12000<br />

9000<br />

6000<br />

3000<br />

Vehicle<br />

Vehicle<br />

MMP1<br />

MMP1<br />

MMP3<br />

MMP buffer<br />

***<br />

MMP3<br />

MMP7<br />

*<br />

*<br />

MMP9<br />

MMP13<br />

MMP buffer<br />

*<br />

MMP7<br />

MMP9<br />

ADAMTS4<br />

ADAMTS4<br />

MMP13<br />

ADAMTS5<br />

ADAMTS5<br />

*<br />

ADAMTS4<br />

94<br />

*<br />

ADAMTS5<br />

NBC2 ng/mL<br />

CTX-II ng/mL<br />

1.6<br />

1.2<br />

0.8<br />

0.4<br />

200<br />

150<br />

100<br />

50<br />

Vehicle<br />

Vehicle<br />

Vehicle<br />

Cysteine buffer<br />

CatK<br />

*<br />

CatB<br />

CatL<br />

Cysteine buffer<br />

CatK<br />

CatB<br />

CatL<br />

Cysteine buffer<br />

Fig. 1. Collagen type II and aggrecan degradation are mediated by different pathways.<br />

Human OA cartilage explants were incubated in MMP and cysteine buffer in the presence or<br />

absence <strong>of</strong> different metalloproteinases [100 ng/ml] and cysteine proteases [100 ng/ml] (Cat =<br />

cathepsin). Biomarkers for collagen type II degradation (CTX-II and NBC2) and aggrecan<br />

degradation (AGNx-II) were measured after 3, 6 and 9h. Data are shown as accumulated for the<br />

three measurements. N=4.<br />

AGNx-II ng/ml<br />

D<br />

F<br />

400<br />

300<br />

200<br />

100<br />

CatK<br />

CatB<br />

CatL<br />

CatS<br />

CatS<br />

CatS


CHAPTER 7: PAPER IV<br />

Detection <strong>of</strong> key proteases to degrade human OA cartilage ex vivo; evaluated by an aggrecan<br />

degradation marker<br />

In human metabolic inactivated OA cartilage incubated in MMP buffer, MMP-3 and -7, and<br />

ADAMTS-4 and -5 all increased aggrecan degradation (AGNx-II) compared to non-stimulated<br />

vehicle (fig. 1E). Addition <strong>of</strong> the MMP inhibitor, GM6001, to the proteases attenuated this<br />

release <strong>of</strong> the aggrecan fragments (data not shown). We did not observe any changes in the<br />

release <strong>of</strong> AGNx-II fragments by MMP-1, -9 and -13, compared to vehicle (fig. 1E).<br />

The cathepsins did not affect the levels <strong>of</strong> AGNx-II significantly; however, we<br />

observed a tendency towards a decrease in the levels <strong>of</strong> AGNx-II compared to vehicle (fig. 1F).<br />

Collagen type II degradation in normal, cytokine-induced, and OA cartilage<br />

In normal bovine cartilage, the endogenous proteinases activated by the MMP-buffer, degraded<br />

the collagen type II in to fragments that were able to be detected by the biomarker, CTX-II. The<br />

MMP inhibitor, GM6001, was able to attenuate CTX-II (Table 1). The bovine cartilage pre-<br />

cultured with cytokines for 4 and 11 days before metabolic inactivated and incubated in MMP<br />

buffer, increased the levels <strong>of</strong> CTX-II compared to normal bovine cartilage, which were also<br />

attenuated in the presence <strong>of</strong> GM6001. However, the human OA cartilage did not generate CTX-<br />

II fragments as in the bovine samples. The cysteine protease inhibitor, E64, did not affect the<br />

CTX-II levels. The biomarker, NBC2, was not detectable in the MMP buffer (Table 1).<br />

The proteinases activated by the cysteine buffer in both bovine and human<br />

cartilage, generated fragments that could be detected by both biomarkers <strong>of</strong> collagen type II<br />

degradation; CTX-II and NBC2 (Table 2). The MMP inhibitor, GM6001 showed a tendency to<br />

decrease CTX-II levels (P=0.057) in normal bovine cartilage, whereas the same inhibitor showed<br />

a tendency to increase CTX-II levels in pre-cultured bovine cartilage (11 days) and in human OA<br />

cartilage (Table 2). Additionally, GM6001 increased NBC2 in human OA and bovine normal and<br />

pre-cultured cartilage. The cysteine protease inhibitor, E64, decreased the levels <strong>of</strong> CTX-II in<br />

human OA and bovine normal and pre-cultured (11 days) cartilage. E64 also decreased the levels<br />

<strong>of</strong> NBC2 in normal bovine cartilage, whereas the cysteine protease inhibitor increased the levels<br />

<strong>of</strong> NBC2 in bovine cartilage pre-cultured with cytokines for 4 days (Table 2). The bovine<br />

cartilage pre-cultured with cytokines for 4 and 11 days before metabolic inactivated and<br />

incubated in cysteine buffer, decreased the levels <strong>of</strong> CTX-II compared to normal bovine cartilage<br />

(Table 2), which was the opposite effects from the one seen in MMP buffer (Table 1).<br />

Furthermore, the human OA cartilage had CTX-II levels corresponding to the cartilage pre-<br />

cultured with cytokines for 11 days (Table 2). Moreover, the levels <strong>of</strong> NBC2 were also lower for<br />

the pre-cultured cartilage (11 days) compared with the normal cartilage. However, the human OA<br />

cartilage had greatly higher levels <strong>of</strong> NBC2 compared to bovine cartilage in general (Table 2).<br />

95


CHAPTER 7: PAPER IV<br />

Aggrecan degradation in normal, cytokine-induced, and OA cartilage<br />

In both bovine and human OA cartilage, the endogenous proteinases activated by MMP-buffer,<br />

degraded the aggrecan in to fragments that were able to be detected by the biomarker, AGNx-II.<br />

The MMP inhibitor, GM6001, was able to attenuate the levels <strong>of</strong> AGNx-II in both bovine and<br />

human OA cartilage (Table 1). The bovine cartilage pre-cultured with cytokines for 4 and 11<br />

days before metabolic inactivation and incubation in MMP buffer, increased the levels <strong>of</strong> AGNx-<br />

II compared to normal bovine cartilage (Table 1). Furthermore, the human OA cartilage had<br />

greatly higher levels <strong>of</strong> AGNx-II than the bovine samples (Table 1). The cysteine protease<br />

inhibitor, E64, did not affect the normal or pre-cultured bovine samples, whereas E64 increased<br />

the levels significantly in the human OA samples (Table 1).<br />

The proteinases activated by the cysteine buffer in both bovine and human cartilage, also<br />

degraded the aggrecan in to fragments that were able to be detected by the biomarker, AGNx-II.<br />

96


CHAPTER 7: PAPER IV<br />

The MMP inhibitor, GM6001, was able to attenuate the levels <strong>of</strong> AGNx-II in normal bovine<br />

cartilage, whereas the pre-cultured bovine cartilage was not affected by the inhibitor (Table 2).<br />

The bovine cartilage pre-cultured with cytokines for 4 and 11 days before metabolic inactivation<br />

and incubation with cysteine buffer, decreased the levels <strong>of</strong> AGNx-II compared to normal<br />

bovine cartilage (Table 2), which was the opposite effects from the one seen in the MMP buffer.<br />

Furthermore, the human OA cartilage had levels <strong>of</strong> AGNx-II as the normal bovine cartilage<br />

(Table 2). The cysteine protease inhibitor, E64, did not affect the normal or pre-cultured bovine<br />

samples, whereas E64 increased the levels significantly in the human OA samples (Table 2).<br />

Discussion<br />

The aim <strong>of</strong> our study was to understand the molecular foundation for the progression <strong>of</strong> articular<br />

cartilage degradation that leads to OA. We analyzed the release <strong>of</strong> cartilage degradation<br />

biomarkers from human OA cartilage by the proteinases reported to play a role in cartilage<br />

degradation 5,10 . Furthermore, we investigated how diseased cartilage is altered from normal<br />

cartilage with regards to the endogenous proteases, and we were observant to possible masking<br />

effects among the proteases.<br />

We showed that the cartilage degradation markers, CTX-II, NBC2, and AGNx-II,<br />

are released in part by different proteolytic pathways in human OA cartilage, as different<br />

proteases generated different biomarkers (fig. 1). The biomarkers were only released by specific<br />

metalloproteinases, and none <strong>of</strong> the supplied cathepsins (fig. 1). Moreover, Cathepsin-B (and a<br />

tendency for Cathepsin-K and Cathepsin-L) inhibited the release <strong>of</strong> CTX-II in cysteine buffer,<br />

suggesting either a protective effect by the cathepsins on cartilage degradation or more likely a<br />

masking effect (fig. 2) from post-cleavage <strong>of</strong> CTX-II fragments by the cathepsins. The aggrecan<br />

degradation increased in the presence <strong>of</strong> both MMPs and aggrecanases, although AGNx-II is<br />

MMP-specific (fig. 1G), suggesting pre- or post-cleavages by the ADAMTS’ may aid the<br />

degradation.<br />

In human OA cartilage explants, the collagen type II and aggrecan may already be<br />

severely degraded by the increased production <strong>of</strong> proteases during the disease 10 , so further<br />

protease-cleavage ex vivo may be difficult to detect, as specific targets measurable by our<br />

biomarkers have already been cleaved and further processed, thereby masking the effect from the<br />

MMPs and cysteine proteases we tested. This possible masking effect could be investigated by a<br />

follow up study, testing the library <strong>of</strong> proteases in normal cartilage and comparing the results<br />

with those from the present study in human OA cartilage.<br />

The endogenous proteases in human OA samples activated in the MMP buffer<br />

(Table 1) failed to increase the levels <strong>of</strong> CTX-II and NBC2. One suggestion could be that the<br />

damaged OA cartilage had less chondrocytes and ECM proteins, and thus fewer enzymes<br />

present, compared to normal cartilage. This is supported by the increased levels <strong>of</strong> CTX-II and<br />

NBC2, when supplying MMP-7 and MMP-9 (fig.1). However, as we were able to decrease MMP-<br />

97


CHAPTER 7: PAPER IV<br />

mediated aggrecan degradation with GM6001 in the MMP buffer, demonstrating that the human<br />

OA cartilage is not depleted for metalloproteinases (table 1).<br />

Fig. 2. Schematic overview <strong>of</strong> the masking effect <strong>of</strong> endogenous<br />

proteases in human OA cartilage. The MMP-mediated neoepitope,<br />

AGNx-II, is further processed by cysteine proteases. Thus,<br />

Inhibition <strong>of</strong> cysteine proteases result in increased AGNx-II.<br />

Several studies by our group have shown that a combination <strong>of</strong> the cytokines, OSM + TNF-α,<br />

induce a damaging effect on cartilage by increasing the production <strong>of</strong> proteases, e.g. aggrecanases<br />

and MMPs 3,4 . This induces a shift from normal turnover towards an uneven turnover - resulting<br />

in excessive protein degradation. In the present study, we pre-stimulated normal bovine cartilage<br />

explants with the cytokines to induce a catabolic system that resemble human OA cartilage<br />

regarding the proteases. This would allow us to have a system representing the proteases in<br />

normal and diseased cartilage, from the same cartilage explant. The cartilage explant pre-<br />

stimulated with cytokines for 4 days increased the aggrecanase-mediated aggrecan degradation,<br />

whereas the cartilage pre-stimulated with cytokines for 11 days increased the MMP-mediated<br />

aggrecan degradation, and not vice versa (data not shown). This confirm that our proteases has<br />

been induced as expected 3 .<br />

98


CHAPTER 7: PAPER IV<br />

The pre-stimulated cartilage incubated in MMP buffer increased the levels <strong>of</strong> CTX-II, and<br />

AGNx-II, suggesting aggrecanases and MMPs to be the key proteases to generate these two<br />

biomarkers. The MMP inhibitor, GM6001, decreased the increased levels <strong>of</strong> the two biomarkers<br />

in both 4 and 11 days pre-stimulated cartilage explants, which correlates with the literature that<br />

GM6001 inhibits both aggrecanases and MMPs 11 . The biomarker levels varied significantly<br />

<strong>between</strong> the normal, cytokine-stimulated and OA cartilage explants, showing that the pre-<br />

stimulated bovine cartilage explants did not represent human OA cartilage. However, the results<br />

informed us instead that the presence and role <strong>of</strong> endogenous proteases vary from healthy<br />

cartilage and during the progression <strong>of</strong> OA. Thus it is <strong>important</strong> to target the right proteases at<br />

the right time, to receive the optimal treatment.<br />

E64 indicated that cysteine proteases mask the levels <strong>of</strong> the biomarker representing<br />

MMP-mediated aggrecan degradation, whereas GM6001 indicated that MMPs masks the levels <strong>of</strong><br />

the collagen type II degradation biomarkers in human OA (Table1/2). Sondergaard et al. 12 have<br />

shown that CTX-II levels increases in the presence <strong>of</strong> E64 in bovine cartilage explants (living<br />

tissue), suggesting that it is a compensatory degrading effect from other proteases, like MMPs,<br />

when cysteine proteases are absent to degrade the proteins. However, it could also be a masking<br />

effect from the cysteine proteases that cleaves MMP-mediated CTX-II fragments further, and<br />

thus masks the levels, which is actually degraded by the MMPs. Our study confirms that the study<br />

by Sondergaard et al. 12 must have been a compensatory effect, as we did not see any increase <strong>of</strong><br />

CTX-II by E64 in cytokine-stimulated bovine explants, in which the chondrocytes were<br />

inactivated. If E64 should have conducted a masking effect in the Sondergaard 12 study, our E64<br />

should have been increased as well.<br />

The present brief report will be followed by a thorough study with regards to the<br />

dynamics <strong>of</strong> proteases in diseased and normal cartilage, and potential compensatory effects <strong>of</strong> the<br />

different proteases herein. A complete understanding <strong>of</strong> the common molecular sequence <strong>of</strong><br />

events underlying the development <strong>of</strong> OA will expand our knowledge <strong>of</strong> the pathogenesis <strong>of</strong><br />

OA. These events will also contribute with <strong>important</strong> information to search for novel therapeutic<br />

targets for the prevention and treatment <strong>of</strong> OA at the right time during the progression <strong>of</strong> the<br />

disease.<br />

Conclusions<br />

Taken together, these experiments suggest that CTX-II, NBC2 and AGNx-II are released in part<br />

by different proteolytic pathways in human OA cartilage. Furthermore, the presence and role <strong>of</strong><br />

endogenous proteases probably depend on the stage <strong>of</strong> OA, as we did not observe the same<br />

release <strong>of</strong> biomarkers during inhibition <strong>of</strong> one type <strong>of</strong> protease, in normal versus cytokine-<br />

stimulated and OA cartilage. Thus more investigation on this topic may have significant value for<br />

development <strong>of</strong> new treatments for OA.<br />

99


References for CHAPTER 7 (PAPER IV)<br />

1 Archer C.W. and Francis-West P. The<br />

chondrocyte. Int J Biochem Cell Biol.<br />

2003;35(4):401-404.<br />

2 Rengel Y., Ospelt C., and Gay S. Proteinases<br />

in the joint: clinical relevance <strong>of</strong> proteinases in<br />

joint destruction. Arthritis Res Ther.<br />

2007;9(5):221-21<br />

3 Madsen S.H., Sumer E.U., Bay-Jensen A.C.,<br />

Sondergaard B.C., Qvist P., and Karsdal M.A.<br />

Aggrecanase- and matrix metalloproteinasemediated<br />

aggrecan degradation is associated<br />

with different molecular characteristics <strong>of</strong><br />

aggrecan and separated in time ex vivo.<br />

Biomarkers. 2010;15(3):266-276.<br />

4 Karsdal M.A., Madsen S.H., Christiansen C.,<br />

Henriksen K., Fosang A.J., and Sondergaard<br />

B.C. Cartilage degradation is fully reversible in<br />

the presence <strong>of</strong> aggrecanase but not matrix<br />

metalloproteinase activity. Arthritis Res Ther.<br />

2008;10(3):R63-<br />

5 Morko J.P., Soderstrom M., Saamanen A.M.,<br />

Salminen H.J., and Vuorio E.I. Up regulation<br />

<strong>of</strong> cathepsin K expression in articular<br />

chondrocytes in a transgenic mouse model for<br />

osteoarthritis. Ann Rheum Dis.<br />

2004;63(6):649-655.<br />

6 Leeming D.J., Bay-Jensen A.C., Vassiliadis E.,<br />

Larsen M.R., Henriksen K., and Karsdal M.A.<br />

Post-translational modifications <strong>of</strong> the<br />

extracellular matrix are key events in cancer<br />

progression: opportunities for biochemical<br />

marker development. Biomarkers.<br />

2011;16(3):193-205.<br />

7 Olsen A.K., Sondergaard B.C., Byrjalsen I.,<br />

Tanko L.B., Christiansen C., Muller A., Hein<br />

G.E. et al. Anabolic and catabolic function <strong>of</strong><br />

chondrocyte ex vivo is reflected by the<br />

metabolic processing <strong>of</strong> type II collagen.<br />

Osteoarthritis Cartilage. 2007;15(3):335-342.<br />

CHAPTER 7: PAPER IV<br />

100<br />

8 Sumer E.U., Sondergaard B.C., Rousseau J.C.,<br />

Delmas P.D., Fosang A.J., Karsdal M.A.,<br />

Christiansen C. et al. MMP and non-MMP<br />

mediated release <strong>of</strong> aggrecan and its fragments<br />

from articular cartilage: a comparative study <strong>of</strong><br />

three different aggrecan and<br />

glycosaminoglycan assays. Osteoarthritis<br />

Cartilage. 2007;15(2):212-221.<br />

9 Hui W., Rowan A.D., Richards C.D., and<br />

Cawston T.E. Oncostatin M in combination<br />

with tumor necrosis factor alpha induces<br />

cartilage damage and matrix metalloproteinase<br />

expression in vitro and in vivo. Arthritis<br />

Rheum. 2003;48(12):3404-3418.<br />

10 Mort J.S. and Billington C.J. Articular cartilage<br />

and changes in arthritis: matrix degradation.<br />

Arthritis Res. 2001;3(6):337-341.<br />

11 Burrage P.S. and Brinckerh<strong>of</strong>f C.E. Molecular<br />

targets in osteoarthritis: metalloproteinases<br />

and their inhibitors. Curr Drug Targets.<br />

2007;8(2):293-303.<br />

12 Sondergaard B.C., Henriksen K., Wulf H.,<br />

Oestergaard S., Schurigt U., Brauer R.,<br />

Danielsen I. et al. Relative contribution <strong>of</strong><br />

matrix metalloprotease and cysteine protease<br />

activities to cytokine-stimulated articular<br />

cartilage degradation. Osteoarthritis Cartilage.<br />

2006;14(8):738-748.<br />

13 Charni-Ben T.N., Desmarais S., Bay-Jensen<br />

A.C., Delaisse J.M., Percival M.D., and<br />

Garnero P. The type II collagen fragments<br />

Helix-II and CTX-II reveal different<br />

enzymatic pathways <strong>of</strong> human cartilage<br />

collagen degradation. Osteoarthritis Cartilage.<br />

2008;16(10):1183-1191.


CHAPTER 8<br />

CHAPTER 8: General discussion and conclusions<br />

General discussion and conclusions<br />

The main aim <strong>of</strong> this PhD project was to investigate the pathogenesis <strong>of</strong> OA with respect to<br />

<strong>bone</strong> and cartilage and the importance <strong>of</strong> the cellular <strong>interactions</strong> <strong>between</strong> these two tissues.<br />

One objective was to develop a pre-clinical model that can be used partly to<br />

investigate the interaction <strong>between</strong> osteoblasts, osteoclasts, and chondrocytes, and partly to test<br />

potential drugs for OA. This objective was successfully achieved by the development <strong>of</strong> the pre-<br />

clinical ex vivo model that included both <strong>subchondral</strong> <strong>bone</strong> and articular cartilage (fig. 13). Femur<br />

heads isolated from 3- to 12-week-old female mice were cultured as explants. These femur heads<br />

comprise a closed system, which are easy to isolate from their host without damaging the<br />

explants. Furthermore, the use <strong>of</strong> different ages <strong>of</strong> mice in the model allowed us to monitor<br />

changes simultaneously in both <strong>bone</strong> and cartilage, as the mice age. Hence, the femur head ex vivo<br />

model has advantages compared to the cartilage ex vivo model (only cartilage). However, the<br />

femur head model also has a disadvantage. Even though we characterized different ages <strong>of</strong> mice,<br />

we were not able to mimic human mature articular cartilage. Mice are short-lived (compared to<br />

human) and always in development, and therefore mimics immature cartilage. However, the<br />

immature cartilage in the murine femur heads contains hypertrophic chondrocytes, which are a<br />

hallmark <strong>of</strong> human OA, suggesting that this system could serve as an <strong>important</strong> disease model. It<br />

is debateable whether the hypertrophic chondrocytes from human OA are the same hypertrophic<br />

chondrocytes we see in the growth plate <strong>of</strong> growing mice.<br />

The established novel ex vivo femur head model was used as a screening model, to<br />

test prednisolone. Our results showed that prednisolone decreased cartilage degradation and<br />

decreased the turnover <strong>of</strong> <strong>bone</strong> (fig. 13). We also tested the effects <strong>of</strong> prednisolone and<br />

dexamethasone on <strong>bone</strong> or cartilage individually in other pre-clinical models, which confirmed<br />

that GCs protect cartilage, but not <strong>bone</strong>. Additionally, GCs did not appear to affect the lifespan<br />

and function <strong>of</strong> resting chondrocyte (fig. 13). These results suggest that the use <strong>of</strong> GC injections<br />

for OA is not preferable without considering the damaging effects on <strong>bone</strong>. Data obtained from<br />

this study also propose that the interaction <strong>between</strong> <strong>bone</strong> and cartilage is <strong>of</strong> less importance in<br />

regard to GC-treatment.<br />

Another objective was to gain greater insight into the proteolytic degradation<br />

processes <strong>of</strong> pathogenic cartilage since cartilage degradation and subsequently loss <strong>of</strong> the tissue is<br />

one <strong>of</strong> the hallmarks <strong>of</strong> the pathogenesis <strong>of</strong> OA. Loss <strong>of</strong> sGAGs from aggrecan is one <strong>of</strong> the first<br />

steps in the pathogenesis <strong>of</strong> OA. We found that there are different molecular structured pools <strong>of</strong><br />

aggrecan that are degraded by different proteases (fig. 13), and we suggest that fully saturated<br />

aggrecan molecules, which are normally found in normal cartilage, are preferentially degraded by<br />

101


CHAPTER 8: General discussion and conclusions<br />

aggrecanases. In contrast, MMPs primarily degrade low saturated aggrecan molecule. This<br />

suggests that it may be favourable to target aggrecanases in early OA and MMPs in later stages <strong>of</strong><br />

OA.<br />

We have also hypothesized that the endogenous proteases found in human OA<br />

depend on the stage <strong>of</strong> disease. This assumption is based on the fact that MMPs, and probably<br />

several other proteases, increase over time during the development <strong>of</strong> OA. However, the<br />

importance <strong>of</strong> cathepsins in this relation is unknown. Thus we used biochemical markers to<br />

assess the different endogenous proteases in normal vs. OA-cartilage. We found that CTX-II,<br />

NBC2 and AGNx-II are released in part by different proteolytic pathways in human OA<br />

cartilage. In addition, we suggest that the presence and role <strong>of</strong> endogenous proteases depend on<br />

the stage <strong>of</strong> OA (fig. 13). Further investigations on this topic may have significant value for<br />

development <strong>of</strong> new treatments for OA.<br />

Figure 13. Schematic illustration summarizing the four papers. The figure shows a femur head divided in a<br />

diseased and a normal half. The development <strong>of</strong> the whole femur head model is seen in PAPER I.<br />

Glucocorticoids (GC) decrease the degradation <strong>of</strong> damaged cartilage, but do not affect normal cartilage. GCs<br />

induce apoptosis in both normal and diseased <strong>bone</strong> (PAPER II). Aggrecan is divided in two different pools, with<br />

different molecular structures, which are degraded by different proteases (PAPER III). Proteases vary <strong>between</strong><br />

normal and OA cartilage. Furthermore, the presence and role <strong>of</strong> endogenous proteases probably depend on the<br />

stage <strong>of</strong> OA (PAPER IV). The figure was produced by Madsen, S.H.<br />

102


Future perspectives<br />

CHAPTER 8: Future perspectives<br />

One <strong>of</strong> the purposes for developing the femur head model was to investigate the cellular<br />

<strong>interactions</strong> <strong>between</strong> <strong>bone</strong> and cartilage, although the GC-study did not provide evidence for<br />

such an interaction <strong>between</strong> the two tissues. Evidently, this needs to be investigated in more<br />

detail in the femur head model. In particular, it is not unlikely that some drugs may affect one<br />

tissue directly, but impinge on another tissue indirectly, i.e., through the other tissue. If a<br />

close coupling <strong>between</strong> cartilage and <strong>bone</strong> turnover exists, future treatment <strong>of</strong> OA most likely<br />

need to restore or rebalance cellular communication in the joint. Otherwise, if the <strong>bone</strong> or<br />

cartilage does not affect one another in the pathogenesis <strong>of</strong> OA, the optimal drug for OA<br />

may be a cocktail <strong>of</strong> tissue-specific drugs.<br />

Future studies could involve the<br />

disassembly <strong>of</strong> the femur head explants in<br />

<strong>bone</strong> and cartilage and compare fractions to<br />

the intact femur head. In this way we may<br />

be able to delineate how the drugs affect<br />

<strong>bone</strong> and cartilage individually as compared<br />

to the entire explants. In this regard, it would be critical to include femur heads from knock-<br />

out mice, which lack the functional role <strong>of</strong> a specific cell type and/or signalling system in<br />

order to investigate cellular crosstalk <strong>between</strong> the two tissues.<br />

In line with our findings in the ex vivo femur head model, it is critical to investigate whether<br />

hypertrophic chondrocytes in this model share similarities to cells in human OA. If the<br />

hypertrophic chondrocytes have the exact same phenotype, it will increase the usefulness <strong>of</strong><br />

the ex vivo model in regard to mimicking human OA cartilage. If we find that the cells are not<br />

exactly alike, we can use the ex vivo model to investigate the hypertrophic chondrocytes we<br />

observe during development.<br />

The present clinical use <strong>of</strong> intra-articular GC injections in OA knees, and the damaging<br />

effects we find on <strong>bone</strong> by GCs in our study, highlights the importance <strong>of</strong> finding a<br />

combination <strong>of</strong> drugs or a new GC, which do not have the detrimental effects on <strong>bone</strong>.<br />

Thus, further studies using the novel femur head model could screen for potential drugs with<br />

no detrimental effect on <strong>bone</strong> or cartilage.<br />

Lastly, it is very <strong>important</strong> to understand the proteolytic processes <strong>of</strong> pathogenic cartilage in<br />

order to developed new treatments for OA, which target the exact proteases that exert the<br />

damaging effects at a given time during the pathogenesis <strong>of</strong> OA. We have already suggested<br />

that it may be favourable to target aggrecanases in early OA and MMPs in later stages <strong>of</strong> OA.<br />

However, more investigation on this topic is needed, especially in regard to investigate other<br />

proteases than aggrecanases and MMPs, e.g., how <strong>important</strong> is the cathepsins in OA?<br />

103


Abbreviations<br />

Abbreviations<br />

342-G2 MMP-mediated 342 FFGV-G2 fragment from aggrecan<br />

374-G2 Aggrecanase-mediated 374 ARGS-G2 fragment from aggrecan<br />

ADAMTS Aggrecanase/A disintergrin and metalloproteinase with thrombospondin motifs<br />

AGNx-II MMP-mediated 342 FFGV-G2 fragment from aggrecan<br />

ALP Alkaline phosphatase<br />

BMP Bone morphogenetic proteins<br />

CC Chondrocyte<br />

COMP Cartilage oligomeric matrix protein<br />

CIIM Collagen type II<br />

CTX-I C-telopeptide <strong>of</strong> type I collagen<br />

CTX-II C-telopeptide <strong>of</strong> type II collagen<br />

DEX Dexamethasone<br />

DMOADs Disease modifying osteoarthritis drugs<br />

ECM Extra-cellular matrix<br />

ELISA Enzyme-linked Immuno Sorbent Assay<br />

E64 Irreversible inhibitor <strong>of</strong> cysteine proteases<br />

FDA The US Food and Drug Administration<br />

G1, G2, G3 Globular domain-1, -2, and -3<br />

GAG Glycosaminoglycan, a proteoglycan<br />

GC Glucocorticoid<br />

GH Growth hormone<br />

GM6001 General MMP inhibitor<br />

GR Glucocorticoid receptor<br />

HA Hyaluronic acid<br />

IGF Insulin-like growth factor-1<br />

IHC Immunohistochemistry<br />

IL Interleukin<br />

mAb Monoclonal antibody<br />

M-CSF Macrophage - colony stimulating factor<br />

MMP Matrix metalloproteinase<br />

MRI Magnetic resonance imaging<br />

NSAIDs Non-steroidal anti-inflammatory drugs<br />

OA Osteoarthritis<br />

OB Osteoblast<br />

OC Osteoclast<br />

ON Overnight<br />

OPG Osteoprotegerin<br />

OSM Oncostatin M<br />

OVX Ovariectomy<br />

PBS Phosphate buffered saline<br />

PICP C-terminal propeptide <strong>of</strong> type I procollagen<br />

PIIANP N-terminal propeptide <strong>of</strong> type II procollagen, splice variant A<br />

PIINP N-terminal propeptide <strong>of</strong> type II procollagen<br />

PINP N-terminal propeptide <strong>of</strong> type I procollagen<br />

PRED Prednisolone<br />

PTH Parathyroid hormone<br />

RA Rheumatoid arthritis<br />

RANK Receptor activator <strong>of</strong> nuclear factor-ĸβ<br />

RANKL Receptor activator <strong>of</strong> nuclear factor-ĸβ ligand<br />

sGAG Sulfated glycosaminoglycans<br />

SD Standard deviation<br />

TGF-β Transforming growth factor β<br />

TIMP Tissue inhibitor <strong>of</strong> metalloproteinase<br />

TNF-α Tumour necrosis factor-α<br />

TRAP Tartrate resistant acid phosphatase<br />

104


Appendix I<br />

Co-author declarations for paper I - IV<br />

Appendix I<br />

105


Appendix I<br />

106


Appendix I<br />

107


Appendix I<br />

108


Appendix I<br />

109


Appendix I<br />

110


Appendix I<br />

111


Appendix II<br />

Publication list<br />

4 first author papers<br />

11 co-Author papers<br />

1 Submitted (first author)<br />

Induction <strong>of</strong> Increased cAMP Levels in Articular<br />

Chondrocytes Blocks Matrix Metalloproteinase–<br />

Mediated Cartilage Degradation, but Not<br />

Aggrecanase-Mediated Cartilage Degradation<br />

Morten Asser Karsdal, Eren Ufuk Sumer, Helle Wulf,<br />

Suzi H. Madsen, Claus Christiansen, Amanda J.<br />

Fosang, and Bodil-Cecilie Sondergaard.<br />

ARTHRITIS & RHEUMATISM, Vol. 56, No. 5,<br />

May 2007, pp 1549–1558.<br />

Patients with Rheumatoid Arthritis have an<br />

altered circulatory Aggrecan Pr<strong>of</strong>ile<br />

Jean Charles Rousseau, Eren U Sumer, Gert Hein,<br />

Bodil C Sondergaard, Suzi H. Madsen, Christian<br />

Pedersen, Thomas Neumann, Andreas Mueller, Per<br />

Qvist, Pierre Delmas and Morten A Karsdal.<br />

BMC Musculoskeletal Disorders, 2008 May 28;9:74.<br />

Cartilage Degradation is Fully Reversible in the<br />

Presence <strong>of</strong> Aggrecanase but not Matrix<br />

Metalloproteinase Activity<br />

Morten Karsdal, Suzi Madsen, Eren Sumer, Claus<br />

Christensen, Kim Henriksen, Amanda Fosang, Bodil<br />

Sondergaard.<br />

Arthritis Res Ther. 2008;10(3):R63. Epub 2008 May 30<br />

Cartilage formation measured by a novel PIINP<br />

assay suggests that IGF-I does not stimulate but<br />

maintains cartilage formation ex vivo<br />

Suzi H. Madsen, Sondergaard BC, Jensen AC,<br />

Morten A Karsdal.<br />

Scand J Rheumatol. 2009 May-Jun;38(3):222-6<br />

Appendix II<br />

112<br />

Biochemical markers and the FDA Critical Path:<br />

how biomarkers may contribute to the<br />

understanding <strong>of</strong> pathophysiology and provide<br />

unique and necessary tools for drug<br />

development.<br />

Karsdal MA, Henriksen K, Leeming DJ, Mitchell P,<br />

Duffin K, Barascuk N, Klickstein L, Aggarwal P,<br />

Nemirovskiy O, Byrjalsen I, Qvist P, Bay-Jensen AC,<br />

Dam EB, Madsen SH, Christiansen C.<br />

Biomarkers. 2009 May;14(3):181-202. Review.<br />

Is <strong>bone</strong> quality associated with collagen age?<br />

Leeming DJ, Henriksen K, Byrjalsen I, Qvist P,<br />

Madsen SH, Garnero P, Karsdal MA.<br />

Osteoporos Int. 2009 Sep;20(9):1461-70.<br />

MAPKs are essential upstream signaling<br />

pathways in proteolytic cartilage degradation -<br />

divergence in pathways leading to aggrecanase<br />

and MMP-mediated articular cartilage<br />

degradation.<br />

Sondergaard BC, Schultz N, Madsen SH, Bay-<br />

Jensen AC, Kassem M, Karsdal MA.<br />

Osteoarthritis Cartilage. 2009 Nov 11. [Epub ahead <strong>of</strong><br />

print]<br />

Suppression <strong>of</strong> MMP activity in bovine cartilage<br />

explants cultures has little if any effect on the<br />

release <strong>of</strong> aggrecanase-derived aggrecan<br />

fragments.<br />

Wang B, Chen P, Jensen AC, Karsdal MA, Madsen<br />

SH, Sondergaard BC, Zheng Q, Qvist P.<br />

BMC Res Notes. 2009 Dec 18;2:259.


Biochemical Markers <strong>of</strong> Joint Tissue Turnover.<br />

Bay-Jensen AC, Sondergaard BC, Christiansen C,<br />

Karsdal MA, Madsen SH, Qvist P.<br />

Assay Drug Dev Technol. 2010 Feb;8(1):118-24<br />

Application <strong>of</strong> biochemical markers in<br />

development <strong>of</strong> drugs for treatment <strong>of</strong><br />

osteoarthritis.<br />

Qvist P, Christiansen C, Karsdal MA, Madsen SH,<br />

Sondergaard BC, Bay-Jensen AC.<br />

Biomarkers. 2010 Feb.;15(1):1-19.<br />

Which elements are involved in reversible and<br />

irreversible cartilage degradation in<br />

osteoarthritis?<br />

Bay-Jensen AC, Hoegh-Madsen S, Dam E,<br />

Henriksen K, Sondergaard BC, Pastoureau P, Qvist<br />

P, Karsdal MA..<br />

Rheumatol Int. 2010 Feb;30(4):435-42.<br />

Aggrecanase- and matrix metalloproteinase-<br />

mediated aggrecan degradation is associated<br />

with different molecular characteristics <strong>of</strong><br />

aggrecan and separated in time ex vivo.<br />

Madsen SH, Sumer EU, Bay-Jensen AC,<br />

Sondergaard BC, Qvist P, Karsdal MA.<br />

Biomarkers. 2010 May 15(3):266-76<br />

Investigation <strong>of</strong> the direct effects <strong>of</strong> salmon<br />

calcitonin on human osteoarthritic chondrocytes.<br />

Sondergaard BC, Madsen SH, Segovia-Silvestre T,<br />

Paulsen SJ, Christiansen T, Pedersen C, Bay-Jensen<br />

AC, Karsdal MA.<br />

BMC Musculoskelet Disord. 2010 Apr. 5;11:62.<br />

Characterization <strong>of</strong> an ex vivo femoral head<br />

model using <strong>bone</strong> and cartilage turnover.<br />

Suzi Hoegh Madsen, Anne S<strong>of</strong>ie Goettrup, Gedske<br />

Thomsen, Søren Tvorup Christensen, Nikolaj<br />

Schultz, Kim Henriksen, Anne-Christine Bay-Jensen,<br />

Morten Asser Karsdal.<br />

Cartilage. 2011 July; vol. 2, 3: pp. 265-278, first<br />

published on October 2010.<br />

Appendix II<br />

113<br />

Glucocorticoids exert context-dependent effects<br />

on cells <strong>of</strong> the joint in vitro.<br />

Suzi Hoegh Madsen, Kim Vietz Andreassen, Søren<br />

Tvorup Christensen, Morten Asser Karsdal, Francis<br />

Michael Sverdrup, Anne-Christine Bay-Jensen, Kim<br />

Henriksen.<br />

Steroids. Acceptet, July 2011<br />

Metalloproteinase and cysteine protease pr<strong>of</strong>iles<br />

in normal and OA cartilage.<br />

Suzi Hoegh Madsen, Kim Henriksen, Søren<br />

Tvorup Christensen, Morten Asser Karsdal, Anne-<br />

Christine Bay-Jensen.<br />

Proteome Science. Submitted, September 2011<br />

Acknowledged for creating figures for papers<br />

Matrix metalloproteinase and aggrecanase<br />

generated aggrecan fragments: implications for<br />

the diagnostics and therapeutics <strong>of</strong> destructive<br />

joint diseases.<br />

Sumer, E. U., Qvist, P. and Tankó, L. B.<br />

Drug Development Research, 2007, 68: 1–13.<br />

Post-translational modifications <strong>of</strong> the<br />

extracellular matrix are key events in cancer<br />

progression: Opportunities for biochemical<br />

marker development.<br />

D. J. Leeming, A. C. Bay-Jensen, E. Vassiliadis, M. R.<br />

Larsen, K. Henriksen, and M.A. Karsdal.<br />

Biomarkers, 2011; 16(3): 193–205.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!