oulu 2019 d 1532 university of oulu p.o. box 8000 fi-90014

106
UNIVERSITATIS OULUENSIS MEDICA ACTA D D 1532 ACTA Jani Luukkonen OULU 2019 D 1532 Jani Luukkonen OSTEOPONTIN AND OSTEOCLASTS IN RHEUMATOID ARTHRITIS AND OSTEOARTHRITIS UNIVERSITY OF OULU GRADUATE SCHOOL; UNIVERSITY OF OULU, FACULTY OF MEDICINE; MEDICAL RESEARCH CENTER OULU

Upload: others

Post on 30-Dec-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

UNIVERSITY OF OULU P .O. Box 8000 F I -90014 UNIVERSITY OF OULU FINLAND

A C T A U N I V E R S I T A T I S O U L U E N S I S

University Lecturer Tuomo Glumoff

University Lecturer Santeri Palviainen

Senior research fellow Jari Juuti

Professor Olli Vuolteenaho

University Lecturer Veli-Matti Ulvinen

Planning Director Pertti Tikkanen

Professor Jari Juga

University Lecturer Anu Soikkeli

Professor Olli Vuolteenaho

Publications Editor Kirsti Nurkkala

ISBN 978-952-62-2363-6 (Paperback)ISBN 978-952-62-2364-3 (PDF)ISSN 0355-3221 (Print)ISSN 1796-2234 (Online)

U N I V E R S I TAT I S O U L U E N S I S

MEDICA

ACTAD

D 1532

AC

TAJani Luukkonen

OULU 2019

D 1532

Jani Luukkonen

OSTEOPONTIN AND OSTEOCLASTSIN RHEUMATOID ARTHRITIS AND OSTEOARTHRITIS

UNIVERSITY OF OULU GRADUATE SCHOOL;UNIVERSITY OF OULU,FACULTY OF MEDICINE;MEDICAL RESEARCH CENTER OULU

Page 2: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014
Page 3: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

ACTA UNIVERS ITAT I S OULUENS I SD M e d i c a 1 5 3 2

JANI LUUKKONEN

OSTEOPONTIN AND OSTEOCLASTS IN RHEUMATOID ARTHRITIS AND OSTEOARTHRITIS

Academic dissertation to be presented with the assent ofthe Doctoral Training Committee of Health andBiosciences of the University of Oulu for public defence inthe Leena Palotie auditorium (101A) of the Faculty ofMedicine (Aapistie 5 A), on 9 November 2019, at 12noon

UNIVERSITY OF OULU, OULU 2019

Page 4: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

Copyright © 2019Acta Univ. Oul. D 1532, 2019

Supervised byProfessor Petri LehenkariProfessor Juha TuukkanenDoctor Sanna Turunen

Reviewed byProfessor Mikko LammiDocent Anna-Marja Säämänen

ISBN 978-952-62-2363-6 (Paperback)ISBN 978-952-62-2364-3 (PDF)

ISSN 0355-3221 (Printed)ISSN 1796-2234 (Online)

Cover DesignRaimo Ahonen

JUVENES PRINTTAMPERE 2019

OpponentProfessor Timothy Arnett

Page 5: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

Luukkonen, Jani, Osteopontin and osteoclasts in rheumatoid arthritis andosteoarthritis. University of Oulu Graduate School; University of Oulu, Faculty of Medicine; MedicalResearch Center OuluActa Univ. Oul. D 1532, 2019University of Oulu, P.O. Box 8000, FI-90014 University of Oulu, Finland

Abstract

Rheumatoid arthritis and osteoarthritis are two chronic joint diseases, which cause two of thelargest socioeconomical burdens among all joint diseases according to the World HealthOrganization. Both diseases are associated with changes in bone structure and bone cell, especiallyosteoclast, function. The etiology or pathogenesis of these diseases are not completely understood.Traditionally, osteoarthritis is seen as a disease resulting from mechanical wear of cartilage andbone, and rheumatoid arthritis as an autoinflammatory disease of synovial tissue. However, alsoin osteoarthritis chronic inflammation is present in synovial tissue, and in rheumatoid arthritislarge changes in bone structure are seen. The field of study focusing on this connection betweeninflammation and bone is called osteoimmunology and it can explain many features of thesechronic diseases linking joint health to disturbances in bone homeostasis.

Here, the study focused on the function of osteoclasts in normal and pathologicalenvironments, and on the factors that have an effect on bone resorption, with a special emphasison the protein osteopontin. Samples of synovial fluid and serum from rheumatoid arthritis andosteoarthritis patients were analyzed for factors affecting osteoclasts, and in vitro cell cultures ofhuman derived osteoclasts were used to analyze osteoclast function in normal and pathologicalenvironment.

The phosphorylation of osteopontin was found to be increased in rheumatoid arthritis, alongwith multiple other inflammatory factors that also affect osteoclasts, such as IL-6, IL-8 and VEGF.Osteoclast cell cultures showed how the use of different patient samples significantly affectedosteoclastogenesis, due to so-called inflammatory osteoclastogenesis. Additionally, we show thatosteoclasts deposit osteopontin into the resorption lacunae during bone resorption.

Based on the results, the inflammatory component present in both osteoarthritis andrheumatoid arthritis significantly affects osteoclast function, and its further study in the future mayreveal new therapeutic possibilities. Especially the new discoveries of osteopontin’s role in normalosteoclast function and its changes seen between osteoarthritis and rheumatoid arthritis may proveto have therapeutic potential.

Keywords: bone resorption, osteoarthritis, osteoclasts, osteoimmunology, osteopontin,rheumatoid arthritis, tartrate-resistant acid phosphatase

Page 6: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014
Page 7: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

Luukkonen, Jani, Osteopontiini ja osteoklastit nivelreumassa ja nivelrikossa. Oulun yliopiston tutkijakoulu; Oulun yliopisto, Lääketieteellinen tiedekunta; Medical ResearchCenter OuluActa Univ. Oul. D 1532, 2019Oulun yliopisto, PL 8000, 90014 Oulun yliopisto

Tiivistelmä

Nivelreuma ja nivelrikko ovat kroonisia nivelsairauksia, jotka Maailman terveysjärjestön(WHO) mukaan aiheuttavat eniten sosioekonomista haittaa. Molemmissa sairauksissa luidenrakenteessa ja luusolujen, erityisesti osteoklastien, toiminnassa tapahtuu muutoksia. Kumman-kaan taudin etiologiaa tai patogeneesiä ei täysin tunneta. Perinteisesti ajatellaan, että nivelrikkojohtuu rusto- ja luukudoksen mekaanisesta kulumisesta ja nivelreuma nivelkalvon autoinflam-matoorisesta tulehduksesta. Kuitenkin nivelrikossa nähdään myös selkeä nivelkalvon krooninentulehdus ja nivelreumassa suuria luun rakenteen muutoksia. Tutkimusala, joka tutkii tulehduk-sen ja luun yhteyttä, on nimeltään osteoimmunologia.

Tässä väitöskirjassa tutkitaan osteoklastien toimintaa ja niihin vaikuttavia tekijöitä, erityises-ti proteiini osteopontiinia, normaalissa ja tautiympäristössä. Analysoin osteoklasteihin vaikutta-via tekijöitä nivelrikko- ja nivelreumapotilaiden näytteistä sekä osteoklastien toimintaa soluvil-jelmissä. Soluviljelmissä käytettiin nivelreuma- ja nivelrikkopotilaiden näytteitä mahdollisim-man totuudenmukaisen ympäristön luomiseksi osteoklasteille.

Tutkimuksessa osoitettiin, kuinka osteopontiinin fosforylaatio on lisääntynyt nivelreumapoti-laiden nivelnesteessä. Myös useiden muiden osteoklasteihin vaikuttavien tekijöiden, kuten IL-6:n, IL-8:n ja VEGF:n, havaittiin lisääntyneen nivelreumassa. Osteoklastien soluviljelmissähavaittiin selkeät erot siinä, miten eri potilasnäytteet vaikuttavat osteoklasteihin ja erityisestitulehduksen aiheuttamaan osteoklastien syntyyn. Osoitan myös, miten osteoklastit erittävätosteopontiinia luunhajotuskuoppaan luun hajotuksen aikana.

Tutkimustulosten mukaan krooninen tulehdustila nivelrikossa ja nivelreumassa vaikuttaahuomattavasti osteoklastien toimintaan. Uskon, että lisätutkimukset tällä saralla voivat paljastaauusia hoidollisia mahdollisuuksia. Erityisesti uudet löydökset osteopontiinin roolista osteoklas-tien toiminnassa sekä muutoksista nivelrikossa ja nivelreumassa vaativat jatkotutkimuksia, jottaproteiinin kliininen merkittävyys saadaan selvitettyä.

Asiasanat: luun hajotus, nivelreuma, nivelrikko, osteoimmunologia, osteoklasti,osteopontiini, tartraatti-resistantti hapan fosfataasi

Page 8: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014
Page 9: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

7

Acknowledgements

This study was carried out at the Department of Anatomy and Cell Biology, Cancer

and Translational Research Unit, Medical Research Center Oulu, University of

Oulu. This study was financially supported by the Emil Aaltonen Foundation and

Oulu Duodecim Society. I owe my deepest gratitude to all the people who have

taught, helped and supported me during these years, and to all the rheumatoid

arthritis and osteoarthritis patients who have given their samples to be studied.

I am deeply grateful to my supervisors Professor Petri Lehenkari, Professor

Juha Tuukkanen and Doctor Sanna Turunen for guidance. I thank Petri and Juha

for allowing me the opportunity to join their research groups as a young medical

student. Petri’s enthusiastic attitude towards medicine and all science in general is

truly remarkable, and he has shown how to combine the careers of an orthopedic

surgeon and a researcher. Juha’s calm supportive positive attitude has been uplifting

at many times of need. Sanna’s enthusiasm towards cells and small details is

astonishing, and without her guidance this thesis would not have reached its best

form.

I also owe thanks to my research colleagues Doctor Elina Kylmäoja and Doctor

Antti Koskela, whose footsteps I have followed on this thesis path. Thanks also to

the rest of Anatomy department’s staff, with special thanks to our laboratory

technicians Miia Vierimaa and Tarja Huhta, whose hands-on guidance and help has

been crucial.

I thank all collaborators and co-authors who took part in this study, with special

thanks to Professor Göran Andersson, and follow-up group Professor Juhana

Leppilahti and Doctor Anna Karjalainen. Professor Mikko Lammi and Docent

Anna-Marja Säämänen are thanked for the careful review of this thesis and the help

to finalize it, and Professor Deborah Kaska for reviewing the English language.

Helka-Maaria Lieslehto is thanked for reviewing the Finnish language.

I am truly honored that Professor Timothy Arnett has accepted the role of

opponent.

My warmest gratitude goes to all my friends from all parts of life. Without you

life would be boring. Thank you for the merry times and adventures in school, work

and leisure activities. May the adventures on and off the mountains never end.

Thanks to my classmates and PhD colleagues Johannes, Joni and Tuomas for

discussions and instructions, your success with your own theses has been a great

motivation.

Page 10: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

8

Finally, I want to thank my family. Johanna and Kalle, I thank you for the love

and support, and raising me to believe that hard work eventually pays off. You laid

the base for everything. Dear Tuija, thank you for believing in me.

12.8.2019 Jani Luukkonen

Page 11: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

9

Abbreviations

ACPAs anti-citrullinated protein antibodies

BMP Bone morphogenetic protein

BMU Basic Multicellular Unit

BSP Bone sialoprotein

CD Cluster of differentiation

CRP C-reactive protein

CTX C-terminal telopeptide

DAMPs damage-associated molecular patterns

DMP1 Dentin matrix protein 1

DSPP Dentin sialophosphoprotein

ELISA Enzyme-linked immunosorbent assay

FE-SEM Field emission scanning electron microscopy

FGF Fibroblast growth factor

G-CSF Granulocyte colony stimulating factor

Gm-CSF Granulocyte-macrophage colony stimulating factor

HSC Hematopoietic stem cell

IFN Interferon

IL Interleukin

IL-1ra Interleukin-1 receptor antagonist

IP-10 Interferon gamma-induced protein 10

MCP-1 Monocyte chemoattractant protein 1

M-CSF Macrophage colony-stimulating factor

M-CSFR Macrophage colony-stimulating factor receptor

MEPE matrix extracellular phosphoglycoprotein

MIP-1-α Macrophage inflammatory protein 1 alpha

MIP-1-β Macrophage inflammatory protein 1 beta

MMP Matrix metalloproteinase

NCP Non-collagenous protein

OA Osteoarthritis

OPG Osteoprotegerin

OPN Osteopontin

PDGF-BB platelet-derived growth factor

PTH Parathyroid hormone

QuOP Cell cycle arrested quiescent osteoclast precursor

RA Rheumatoid arthritis

Page 12: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

10

RANK Nuclear factor kappa-Β

RANKL Nuclear factor kappa-Β ligand

RF rheumatoid factor

Runx2 Runt-related transcription factor

SIBLING Small Integrin-Binding Ligand, N-linked Glycoprotein

Th T helper cell

TNF-α Tumor necrosis factor-alpha

TRAcP Tartrate-resistant acid phosphatase

VEGF Vascular endothelial growth factor

WHO World Health Organization

Wnt Wingless and integration

αvβ3 Alpha-v beta-3 integrin

Page 13: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

11

List of original publications

This thesis is based on the following publications, which are referred to throughout

the text by their Roman numerals:

I Luukkonen, J., Pascual, L. M., Patlaka, C., Lång, P., Turunen, S., Halleen, J., Nousiainen, T., Valkealahti, M., Tuukkanen, J., Andersson, G., Lehenkari, P. (2017). Increased amount of phosphorylated proinflammatory osteopontin in rheumatoid arthritis synovia is associated to decreased tartrate-resistant acid phosphatase 5B/5A ratio. Plos One, 12(8). doi:10.1371/journal.pone.0182904.

II Luukkonen, J., Hilli, M., Nakamura, M., Ritamo, I., Valmu, L., Kauppinen, K., Tuukkanen J., Lehenkari, P. (2019). Osteoclasts secrete osteopontin into resorption lacunae during bone resorption. Histochemistry and Cell Biology. doi:10.1007/s00418-019-01770-y.

III Luukkonen J., Huhtakangas J., Turunen S., Tuukkanen J., Vuolteenaho O., Lehenkari P. (2019). Bone resorption and osteoclastogenesis are stimulated by synovial fluid from osteoarthritis and rheumatoid arthritis patients in vitro. Manuscript.

Page 14: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

12

Page 15: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

13

Contents

Abstract

Tiivistelmä

Acknowledgements 7 

Abbreviations 9 

List of original publications 11 

Contents 13 

1  Introduction 15 

2  Review of the literature 17 

2.1  Bone ........................................................................................................ 17 

2.1.1  Bone matrix .................................................................................. 19 

2.1.2  Bone cells ..................................................................................... 21 

2.1.3  Osteoclasts and bone resorption ................................................... 24 

2.1.4  Bone remodeling .......................................................................... 30 

2.2  Joint ......................................................................................................... 32 

2.3  Osteoimmunology ................................................................................... 33 

2.4  Osteopontin (OPN) ................................................................................. 35 

2.5  Tartrate-resistant acid phosphatase (TRAcP) .......................................... 38 

2.6  Rheumatoid arthritis (RA) ...................................................................... 41 

2.6.1  Osteoimmunology in RA .............................................................. 45 

2.7  Osteoarthritis (OA) ................................................................................. 47 

2.7.1  Osteoimmunology in OA ............................................................. 50 

3  Aims of the study 53 

4  Materials and methods 55 

5  Results 57 

5.1  Analyses of OPN and TRAcP in RA synovial fluid and tissue (I) .......... 57 

5.1.1  Phosphorylation of OPN in synovial fluid (I) .............................. 57 

5.1.2  TRAcP 5A and 5B concentrations in synovial fluid (I) ................ 58 

5.1.3  Localization of OPN and TRAcP in RA and OA patient

synovial tissue (I) ......................................................................... 58 

5.2  Analyses of OPN in resorption lacunae (II) ............................................ 60 

5.2.1  Detection of OPN in resorption lacunae using FE-SEM

(II) ................................................................................................ 60 

5.2.2  Analysis of proteins enriched in the resorbed bone slices

(II) ................................................................................................ 61 

5.2.3  Analysis of glycan epitope in the resorption lacunae (II) ............. 61 

Page 16: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

14

5.3  Immunohistology of OPN and TRAcP in the areas of increased

bone metabolism in OA (II) .................................................................... 62 

5.4  Analyses of the effect of RA serum and synovial fluid on

osteoclastogenesis and bone resorption (III) ........................................... 64 

5.4.1  Effect of novel untreated RA patient serum on

osteoclastogenesis and bone resorption ........................................ 65 

5.4.2  Effect of RA and OA patients’ synovial fluid on

osteoclastogenesis and bone resorption ........................................ 66 

5.4.3  Cytokine analyses of the patient samples ..................................... 66 

6  Discussion 69 

6.1  Decreased TRAcP 5B/5A ratio leads to increased

phosphorylation of OPN in RA synovial fluid. ....................................... 69 

6.2  OPN is one of the key proteins secreted into the resorption

lacunae during bone resorption ............................................................... 71 

6.3  OPN is found in the calcified cartilage ...................................................... 74 

6.4  RA synovial fluid and serum increase osteoclastogenesis and

bone resorption in vitro ........................................................................... 74 

6.5  Future aspects .......................................................................................... 78 

7  Conclusions 81 

List of references 83 

Original publications 101 

Page 17: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

15

1 Introduction

Bones give humans and other vertebrates their distinct appearance and form. Other

tissues and organs have their own specific place within or outside the skeleton. The

bones are connected to each other by joints, which allows us to move.

For a long time, bone tissue was considered only as an inert tissue functioning

as a scaffold supporting and protecting other tissues and vital organs. However later,

based on the advances in research of human physiology, it was discovered that bone

is a living and constantly changing tissue, which functions as the body’s primary

reservoir of calcium and phosphate ions and serves many other vital functions, such

as hematopoiesis.

During the last decades, osteoimmunology, bone’s function in regulating the

body’s immunological processes, has been a topic of high interest. Recent advances

in the field have shown how the bone tissue immune-system connection plays a

part in the pathological process of many chronic diseases, such as rheumatoid

arthritis (RA), osteoarthritis (OA) or osteoporosis (Okamoto & Takayanagi, 2019).

The discoveries have led to the rapid development of drugs that effect the bone

tissue immune-system interaction, such as denosumab or adalimumab. However,

much work is still needed to gain a deeper understanding in this relatively new field

of study.

According to the World Health Organization (WHO), two chronic rheumatic

conditions that cause the greatest impact on society are RA and OA (Briggs et al.,

2018). The prevalence of both diseases is on the rise, and they cause a huge amount

of morbidity and disability to the affected individuals and a significant economic

burden on societies throughout the world. Unfortunately, the pathogeneses of these

diseases are not completely understood and treatments even with modern advances

are limited.

RA and OA are both diseases characterized by typical pathological changes in

bone metabolism, local bone loss and synovitis. This study was conducted to

examine the osteoimmunological mechanisms, especially the ones leading to bone

loss, in the two diseases, and osteoclast function in disease and normal

environments. I focus on how the diseases affect osteoclastogenesis and possible

mechanisms behind it, with a special emphasis on osteopontin (OPN) protein.

Page 18: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

16

Page 19: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

17

2 Review of the literature

2.1 Bone

The human skeleton consists of around 270 bones at the time of birth. During

growth multiple bones fuse together and therefore the number of bones in an adult

skeleton is around 206 (Gray, 1918). The main function of the skeleton is to act as

a scaffold and to protect other tissues of the human body. Other vital functions of

bone tissue are hematopoiesis in bone marrow and serving as the body’s calcium

and phosphate reservoir.

Based on a bone’s gross appearance two forms of bone can be distinguished,

compact bone and trabecular bone. To the naked eye a cross section of compact

bone appears as a solid mass and trabecular bone as a network of thin bone

trabeculae. Generally compact bone is found at the shaft, diaphysis, of long bones,

such as the femur, where it forms a hollow cylinder with the central cavity called

the bone marrow cavity. The ends of long bones, metaphysis and epiphysis, are

made of trabecular bone, which is covered by a thin layer of compact bone.

(Kierszenbaum & Tres, 2015)

Under a microscope, bone can be divided into two types based on the

organization of the extracellular matrix: lamellar bone, typical in mature bone, and

woven bone, typical in developing bone. In typical compact lamellar bone, the

organized bone matrix is seen as layers, lamellae, surrounding a Haversian or

Volkmann’s canal. Haversian canals are longitudinal blood vessel channels within

the compact bone tissue. Volkmann’s channels are similar but transverse or oblique

and can connect the Haversian channels. The combination of the lamellae and the

canal is called an osteon, which is separated from other osteons by a cement line

and interstitial lamella. In trabecular bone the lamellae are parallel to the trabecular

surface, as there are no blood vessel channels and the bone cells receive nutrients

directly from the bone marrow. The anatomical structures of bone are presented in

figure 1. (Kierszenbaum & Tres, 2015; Repp et al., 2017; Weatherholt, Fuchs, &

Warden, 2011)

Page 20: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

18

Fig. 1. The architecture of a long bone. Anatomically a bone can be divided into three

sections, diaphysis, metaphysis and epiphysis, and by macroscopic appearance into

compact and trabecular bone. Diaphysis is the shaft of a long bone. Metaphysis and

diaphysis are divided by the growth or epiphyseal plate during growth, and in adults by

the epiphyseal line after the cartilage in the growth plate turns to bone. Trabecular bone

is sponge-like bone present at the ends of a long bone, which is covered by a thin layer

of compact bone. The diaphysis is composed only of compact bone at the perimeter

and a hollow bone marrow cavity in the middle. Microscopically, trabecular bone is

made of small bone trabeculae, which are surrounded by bone marrow. In compact

bone, osteons and Haversian channels can be seen with a microscope. The bone is

surrounded by periosteum on the outer surface and by endosteum on the inner surface.

Modified from Kierszenbaum et al., 2015 and Weatherholt, Fuchs, & Warden, 2011.

Bone tissue consists of extracellular matrix (90%) and bone cells (10%) (Mansour,

Mezour, Badran, & Tamimi, 2017). The extracellular matrix is composed of

inorganic (65%) and organic parts (25%), and water (10%) (Boskey, 2013). The

bone tissue is a live tissue, which is constantly degraded and rebuilt by bone cells.

The process is called bone remodeling or bone metabolism. In adults it should be

in equilibrium. A disturbance in the process can lead to the onset of diseases, such

as osteoporosis when bone degradation is increased or osteopetrosis when bone

deposition is increased pathologically (Hadhidakis & Androulakis, 2006).

Page 21: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

19

2.1.1 Bone matrix

Ninety percent of bone is extracellular matrix which is 65% inorganic mineral,

mostly carbonated hydroxyapatite (Ca10(PO4)6(OH)2); the organic part

constitutes roughly around 25% of the extracellular matrix and the rest is water

(Boskey, 2013). The organic part is made of mostly type I collagen (90%) and small

amounts of collagen type III and V, non-collagenous proteins (NCPs) (8%) and

lipids (2%) (Young, 2003).

Bone mineral density is the main component of bone’s strength and stiffness

(Seeman & Delmas, 2006). Carbonated hydroxyapatite is the main mineral

substrate in bone and the basis of bone’s stiffness. It forms hexagonal crystals in

the scaffold created by collagen fibers and NCPs to form the rigid bone matrix

(Florencio-Silva, Sasso, Sasso-Cerri, Simões, & Cerri, 2015). Along with

hydroxyapatite the bone mineral matrix contains also significant amounts of

bicarbonate, sodium, potassium, citrate, magnesium, carbonate, fluoride, zinc,

barium, and strontium, which also affect the mechanical properties of the bone

matrix (Florencio-Silva et al., 2015). The correct bone mineral density is important

for the bone function, if the mineral density is too low the bones are weak, but if

the mineral density is too high the bone loses its elasticity and becomes brittle

(Seeman & Delmas, 2006). Also, the bone mineral density varies between species

and bones depending on their function. For example, the stapes of the human ear is

98% mineral so it is very stiff and can transfer sound better, whereas in the average

human bone the mineral density is around 60% to make it more elastic and damage

resistant (Boskey, 2013).

To create the scaffold for the bone minerals, collagen type I, which is produced

by the bone forming cells, osteoblasts, self assembles into a triple helix form (three

separate molecules, two alpha-1 chains and a one alpha-2 chains) and groups triple

helices form a microfibril via covalent cross linking (Stock, 2015). Multiple fibrils

come together to form a fiber. Water stabilizes the collagen structure by forming

hydrogen bonds between the fibers (Stock, 2015). If the fibril formation is

interrupted or faulty, the scaffold will not be able to stay together and hold the

hydroxyapatite, as can be seen in a genetic disease osteogenesis imperfecta (Young,

2003). In certain types of osteoporosis, a faulty collagen scaffold is also thought be

a factor (Young, 2003).

Page 22: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

20

Non-collagenous proteins

Bone tissue contains various non-collagenous proteins (NCPs) produced by

different bone cells among its collagen fibers. Most of the NCPS are glycoproteins

or proteoglycans, either located at the bone cell surfaces or in the extracellular

matrix (Young, 2003). The NCPs exert various important functions in the bone, e.g.,

they bind into the collagen fibers and hydroxyapatite and take part in the

mineralization process, act as paracrine signal molecules in bone, or as cytokines

for inflammatory cells in bone or further away in other tissues as endocrine

molecules (Florencio-Silva et al., 2015; Gramoun et al., 2010). Most NCPs are not

bone specific proteins and their expression is found throughout the human body

which, along with the fact that many of them have multiple overlapping functions

also in bone, has made them hard to study and their functions are not thoroughly

understood (Morgan, Poundarik, & Vashishth, 2015; Young, 2003).

An interesting group of bone NCPs are the SIBLING proteins (Small Integrin-

Binding Ligand, N-linked Glycoprotein), which all contain an RGD-sequence

(Arg-Gly-Asp). Integrin receptors found at the cell surfaces of, e.g., osteoclasts and

osteoblasts, can bind the RGD sequence (Qin, Baba, & Butler, 2004). Proteins that

belong in this group are osteopontin (OPN), bone sialoprotein (BSP), dentin matrix

protein 1 (DMP1), dentin sialophosphoprotein (DSPP), and matrix extracellular

phosphoglycoprotein (MEPE). Common for these proteins is that they have

multiple post-translational modifications, such as phosphorylation, glycosylation,

and proteolytic processing, which have significant effects on their structure and

biological functions (Qin et al., 2004). The various modifications present in the

proteins has made it hard to solve their various distinct functions. The proteins were

originally identified in bone, and accordingly have their particular roles in

biomineralization and bone metabolism, but many of them have also been found to

play various interesting roles in other human tissues. OPN, for example, is an

important mediator of inflammation, but also has been detected in various cancers

(Morimoto, Kon, Matsui, & Uede, 2010; Pietras et al., 2014).

Other NCPs of interest in bone are fibronectin, osteonectin, osteocalcin and

sclerostin. Fibronectin is one of the most abundant NCPs in bone and also contains

the same RGD sequence as the SIBLING proteins (Young, 2003). Fibronectin

accumulates around sites of new bone formation, where it likely acts as an

attachment point for bone cells and regulates especially osteoblast function and

bone mineralization (Young, 2003). Osteonectin is another protein that is found in

large quantities in bone; it is believed to act as a connector molecule between the

Page 23: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

21

mineral crystals and collagen, and also to take part in regulating osteoblast and

osteoclast activity (Rosset & Bradshaw, 2016). Osteocalcin is an interesting NCP

produced by osteoblasts. Its primary biological functions are actually outside of

bone, where it has been shown to play only a small role in regulating bone mineral

density and its main functions are thought to be as an endocrine molecule that

regulates the body’s glucose homeostasis, brain development, cognition, and male

fertility (Moser & van, 2019). Sclerostin is a natural inhibitor of osteoblast function

and bone formation produced by osteocytes. This discovery has led to the rapid

research and development of new promising osteoporosis drugs (Bhattacharyya,

Pal, & Chattopadhyay, 2018).

Along with the few NCPs mentioned above, various other NCPs can also be

found in bone, for example, bone morphogenetic proteins (BMPs), various growth

factors and small leucine-rich proteoglycans, such as decorin, biglycan, lumican

and osteoaderin etc., many of their biological functions or clinical relevance are

still unknown (Florencio-Silva et al., 2015).

2.1.2 Bone cells

Bone cells make up 10% of bone tissue (Mansour et al., 2017). The bone cells come

from two different lineages; the osteoprogenitor cells, osteoblasts and osteocytes,

are of mesenchymal origin and the osteoclasts derive from the monocyte-

macrophage line of hematopoiesis (Kierszenbaum & Tres, 2015). In general,

osteoblasts are bone forming cells, osteoclasts resorb bone and osteocytes control

the bone metabolism.

Osteoblasts

The osteoblasts account for 4-6% of all bone cells (Capulli, Paone, & Rucci, 2014).

They are derived from the mesenchymal stem cells residing near bone surfaces,

which first evolve into osteoprogenitor cells, and then into mature osteoblasts

capable of bone formation (Roeder, Matthews, & Kalajzic, 2016). The process is

controlled by various cytokines and BMPs. During osteoblast maturation specific

signaling pathways are activated: the three most important and specific for

osteoblastogenesis are Wnt (Wingless and integration), Runx2 (Runt-related

transcription factor) and osterix (Sp7) (Roeder et al., 2016). Knockout-out mice

without these genes exhibit severely decreased bone formation due to impaired

osteoblastogenesis (Roeder et al., 2016).

Page 24: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

22

During bone formation the mature osteoblasts create the organic matrix which

is then mineralized. The osteoblasts secrete the collagens, mostly type I collagen,

and multiple NCPs, such as osteocalcin, osteonectin, BSP and OPN, to create the

organic scaffold, which is then mineralized (Capulli et al., 2014; Roeder et al.,

2016). During the mineralization, osteoblasts produce alkaline phosphatase, which

degrades the mineralization inhibitor pyrophosphate, and thus allows

mineralization and secretion of matrix vesicles containing hydroxyapatite crystals

that are incorporated into the extracellular organic matrix (Capulli et al., 2014;

Roeder et al., 2016). After actively forming the bone matrix the osteoblasts can

undergo apoptosis, become embedded in the matrix and differentiate into

osteocytes, or become quiescent bone lining cells depending on their location and

other bone cells present.

Osteoblasts have various other functions besides bone formation. Particularly

they control osteoclastogenesis and bone resorption by secreting three of the most

important cytokines that affect osteoclast differentiation and activation; receptor

activator of nuclear factor kappa-Β ligand (RANKL), macrophage colony-

stimulating factor (M-CSF) and osteoprotegerin (OPG) (Sato et al., 2013).

Osteoblasts also affect processes outside of bone by secreting osteocalcin, which

acts as an endocrine molecule as explained earlier (Moser & van, 2019), and

participate in the regulation of the hematopoietic system by regulating

hematopoietic stem cell homing, mobilization and number, and especially B-cell

lymphopoiesis (Kode et al., 2014).

Osteocytes

Osteocytes are spider shaped cells embedded inside the bone matrix, which account

for 90-95% of all bone cells (Capulli et al., 2014). The area around the cell body is

called a lacuna, and the osteocytes are connected to each other via canaliculi, where

adjacent cells are connected to each other by gap junctions. Via this lacuna-

canalicular system oxygen and nutrients are transported to the cells living in the

encapsulated environment (H. Chen, Senda, & Kubo, 2015). During the

transformation process from an osteoblast to an osteocyte the cell volume and

number of cell organelles are considerably decreased. The expression of key

osteoblast proteins is decreased or stops and the expression of key osteocyte

proteins begins. Such proteins have been identified to include DMP1, MEPE,

fibroblast growth factor (FGF), dickkopf-related protein 1 and sclerostin, which

have been shown to affect bone formation and mineralization and phosphate

Page 25: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

23

metabolism (H. Chen et al., 2015). Of these proteins, especially sclerostin has been

of great interest recently. Sclerostin inhibits the Wnt signaling pathway in the

osteoblasts, which decreases bone formation. Inhibition of sclerostin’s action with

anti-sclerostin drugs have shown promise as new osteoporosis treatments in animal

models and clinical trials are currently being conducted (Bhattacharyya et al., 2018).

The main function of an osteocyte is to act as a regulator of bone metabolism

by regulating both osteoclasts and osteoblasts (Florencio-Silva et al., 2015).

Osteocytes act as mechanoreceptors in bone. They sense load and possible fractures

or other changes, and transmit signals to other bone cells via the lacuna-canalicular

system (Capulli et al., 2014; Florencio-Silva et al., 2015). Mechanical stress

increases bone mass and strength, while rest decreases them (Robling & Turner,

2009). The regulation of bone metabolism by osteocytes is not completely

understood, but it seems that osteocyte apoptosis under load is one crucial factor

(Capulli et al., 2014; H. Chen et al., 2015). Too little or too great mechanical

stimulus causes osteocyte apoptosis (Robling & Turner, 2009). The apoptotic

osteocytes release various cytokines, most importantly RANKL, tumor necrosis

factor-alpha (TNF-α), interleukin-6 (IL-6) and IL-11 (Capulli et al., 2014; H. Chen

et al., 2015). The cytokines cause an increase in osteoclast differentiation and

activity resulting in increased bone turnover (Amarasekara et al., 2018). There is

controversial evidence as to whether osteocyte apoptosis decreases or increases

osteoblast activity (Capulli et al., 2014; H. Chen et al., 2015). However, the

evidence that bone mass and strength increase under load is consistent (Robling &

Turner, 2009). It has been suggested that under load sclerostin production in

osteocytes is decreased, which results in increased osteoblast activity (Bullock,

Pavalko & Robling, 2019). Further research is needed on this subject before

conclusions can be drawn on how osteocytes regulate osteoblast function, but the

consensus seems to be that osteocytes are the link between osteoclast and osteoblast

action (Capulli et al., 2014; H. Chen et al., 2015).

Osteocytes also have effects outside the bone. They function as endocrine cells

by secreting FGF-23, which controls phosphate and D-vitamin metabolism by

affecting kidney cells (Bonewald & Wacker, 2013). The osteocytes have also been

shown to have immune- and fat metabolism regulatory functions, as osteocyte

deficient mice had severe lymphopenia due to impaired T- and B-cell

lymphopoiesis and a total lack of white adipose tissue (Sato et al., 2013).

Page 26: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

24

2.1.3 Osteoclasts and bone resorption

Osteoclasts are defined as multinucleated tartrate-resistant acid phosphatase

(TRAcP) expressing giant cells capable of resorbing mineralized bone (Pierce,

1991). They differentiate from the monocyte-macrophage line of hematopoiesis by

fusing from mononuclear precursor cells into multinucleated TRAcP positive cells

(Boyle, Simonet, & Lacey, 2003). Most common human bone diseases are due to

excess osteoclast activity, which is a result of an imbalance of factors regulating

bone metabolism (Rodan & Martin, 2000). Such diseases include osteoporosis,

osteoarthritis, rheumatoid arthritis, multiple myeloma and metastatic cancers.

Like the other bone cells, the osteoblasts and the osteocytes, the osteoclasts

also have various functions other than just their primary job as bone degraders. The

osteoclasts take part in the regulation of bone metabolism by affecting the

osteoblasts, as well as the regulation of hematopoiesis and angiogenesis in bone

(Charles & Aliprantis, 2014). The osteoclasts also have important immunological

functions. The term osteoimmunology was introduced around the turn of the

millennium, almost twenty years ago now, when osteoclasts and immune cells,

especially T-cells, were found to have a tight relationship regulating each other’s

function (Arron & Choi, 2000; Takayanagi et al., 2000).

Osteoclastogenesis

During osteoclastogenesis, monocytes from the myeloid line of hematopoiesis fuse

together to create the osteoclasts. The pre-osteoclast cells capable of fusing together

have been suggested to be cells of the monocyte linage expressing cluster of

differentiation (CD) molecule CD14 (Costa-Rodrigues, Fernandes, & Fernandes,

2011; Hemingway et al., 2011; Sørensen et al., 2007). In the presence of M-CSF

the cells start expressing RANK, a signaling receptor for RANKL, and this

activation is the main promoter of differentiation and fusion of pre-osteoclasts into

osteoclasts (Park-Min, 2018).

Osteoclast precursor cells can be found in bone marrow and circulating in

peripheral blood. It has been suggested that after initial differentiation from

monocytes to pre-osteoclasts in the bone marrow or in another hematopoietic organ

the cells undergo cell cycle arrest and circulate in the blood stream as cell cycle

arrested quiescent osteoclast precursors (QuOPs) (Kotani et al., 2013; Mizoguchi

et al., 2009). The QuOPs circulate in the blood until they eventually reach the bone

surfaces, where they undergo the final steps of osteoclastogenesis by fusing (Kotani

Page 27: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

25

et al., 2013; Mizoguchi et al., 2009). Figure 2 presents a schematic presentation of

osteoclast precursor differentiation and homing.

Fig. 2. Osteoclast precursor differentiation and homing. The pre-osteoclasts proliferate

and differentiate from progenitor cells of the monocyte line in hematopoietic organs,

such as bone marrow and spleen. After differentiation, the pre-osteoclasts undergo cell

cycle arrest then circulate in the blood stream and home towards bone, where they

undergo fusion and differentiation into the osteoclasts in the presence of

osteoclastogenesis promoting factors, such as M-CSF and RANKL produced by the

osteoblasts and the osteocytes.

The generation of the osteoclasts from the pre-osteoclasts is most importantly

controlled by two cytokines M-CSF and RANKL, which both are produced by cells

of the osteoblast lineage, the osteoblasts and the osteocytes, of which the

osteoblasts are thought to be more important producers of osteoclastogenesis

regulating factors (Sato et al., 2013). First M-CSF signaling activates Akt and ERK

signal pathways inside the pre-osteoclasts, which causes an increase of cell surface

receptor RANK production and induction of the main osteoclast marker TRAcP

production (Amarasekara et al., 2018; Park-Min, 2018). RANKL activates the

RANK receptor and causes multiple other signaling pathways to be induced in the

pre-osteoclast and production of the key osteoclast proteins, which promote cell

fusions and the final differentiation into the mature osteoclasts (Amarasekara et al.,

2018; Park-Min, 2018). RANKL is the most important factor of osteoclastogenesis,

Page 28: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

26

as it has been shown in knockout mice models that in the absence of either RANK

or RANKL osteoclastogenesis does not happen (Dougall et al., 1999; Kong et al.,

1999; J. Li et al., 2000). During the last stage of osteoclast differentiation,

production of proteins involved in the adhesion of the cell to the bone matrix and

resorption activity, such as alpha-v beta-3 integrin (αvβ3) and cathepsin K, begins

(Boyle et al., 2003; Soysa & Alles, 2016). The cells attach themselves to the bone

matrix and become polarized as their cytoskeleton is reshaped creating the unique

morphology of an osteoclast (Boyle et al., 2003; Soysa & Alles, 2016).

As explained earlier in chapter 2.1.2, osteocytes act as mechanosensors in bone

and produce signals in areas where increased bone turnover is needed. Therefore,

it has been suggested that they control the location of osteoclast activation.

However, the specific mechanisms are as yet unknown, but osteocyte apoptosis has

been suggested to be key factor as it causes production of osteoclastogenesis

promoting factors, such as RANKL, in the surrounding viable osteocytses (Capulli

et al., 2014; H. Chen et al., 2015).

The main controlling cell of osteoclastogenesis is the osteoblast, which along

with the production of M-CSF and RANKL also produces OPG, a competitive

inhibitor of RANKL, which is the most important inhibitor of osteoclastogenesis

(Park-Min, 2018). OPG knockout mice suffer from severe osteoporosis, and

transgenic mice overexpressing OPG suffer from osteopetrosis (Bucay et al., 1998;

Simonet et al., 1997). Reciprocally osteoclasts also affect osteoblast function by

secreting so-called clastokines, which either increase or decrease osteoblast activity

(Cappariello, Maurizi, Veeriah, & Teti, 2014). Clastokines that are believed to

increase osteoblast activity include TRAcP, sphingosine-1-phosphate, BMP6,

wingless-type 10b, hepatocyte growth factor and collagen triple helix repeat-

containing protein 1, and clastokines thought to decrease osteoblast activity include

platelet derived growth factor BB (PDGF BB) and sclerostin (Teti, 2013). However,

the effect of osteoclasts on osteoblasts is not as well understood as the osteoblasts’

effect on the osteoclasts. Further studies are needed to gain a full understanding of

the osteoclast-osteoblast connection and effect on the bone metabolism.

Various other factors which influence osteoclastogenesis less significantly than

M-CSF, RANKL and OPG are also known. Various inflammatory cytokines have

been shown to increase osteoclastogenesis either by the RANKL pathway or

independently. These include TNF-α, IL-1, IL-6, IL-7, IL-8, IL-11, IL-15, IL-17,

IL-23, IL-34, C-reactive protein (CRP) and vascular endothelial growth factor

(VEGF) (Amarasekara et al., 2018; H. Kim, Kim, Kim, Cho, & Lee, 2015; K. Kim,

Kim, Moon, Lee, & Kim, 2015). Reciprocally various cytokines are also shown to

Page 29: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

27

decrease osteoclastogenesis, which include interferon-α (IFN-α), IFN-β, IFN-γ, IL-

3, IL-4, IL-10, IL-12, Il-27 and IL-33 (Amarasekara et al., 2018). Increased

inflammatory osteoclastogenesis can be seen in inflammatory diseases, such as RA,

but interestingly the phenotype of inflammatory osteoclasts has been found to differ

from the normal osteoclast. The inflammatory osteoclasts are described to be

smaller and contain fewer nuclei, and thus present a more macrophage like

phenotype as a result of a strong inflammatory stimulus (Suzuki et al., 2001; Tsuboi

et al., 2003).

Important physiological factors that regulate osteoclastogenesis are pH and

hypoxia. Low pH and oxygen levels have been shown to increase

osteoclastogenesis (Arnett, 2010; Yuan et al., 2016). The optimal pH for the

osteoclasts has been suggested to be around 6.8 (Arnett, 2010; Yuan et al., 2016),

while the normal arterial blood pH in the human body is around 7.4. In normal

tissue the interstitial oxygen pressure is at a minimum of around 3%, but optimal

osteoclastogenesis has been shown to occur in 1-2% oxygen (Arnett, 2010).

Bone resorption

To resorb bone the mature osteoclasts must first attach themselves to the matrix and

become polarized. First the osteoclast forms a sealing zone, a structure which

attaches the osteoclasts to the extracellular matrix (Takito, Inoue, & Nakamura,

2018). As the sealing zone is formed the osteoclast becomes polarized. The cell

membrane area inside the sealing zone is called a ruffled border membrane and the

cell membrane on the opposite side of the cell is called the functional secretory

domain.

The sealing zone and the ruffled border are formed as the cell attaches to the

matrix with multiple podosomes, foot-like short cellular protrusions present at the

cell membrane (Rucci & Teti, 2016). Each podosome is made of an actin

microfilament core and integrins that protrude outside of the cell (Rucci & Teti,

2016; Takito et al., 2018). The αVβ3 integrin has been suggested to be the main

integrin facilitating osteoclast attachment, as it recognizes the RGD amino acid

sequence present in various bone NCPs, such as fibronectin, vitronectin and the

SIBLING proteins (OPN, BS, DMP1, DSPP, MEPE) (Qin et al., 2004; Ross et al.,

1993). The round sealing zone completely segregates the extracellular area between

the osteoclast and the resorbed matrix from the rest of the environment. This

microenvironment is called the resorption lacuna or pit. The origin of the proteins

to which the αVβ3 integrin attaches is not fully understood. There are proteins

Page 30: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

28

already present in bone, but the osteoclasts have also been suggested to produce

them (Dodds et al., 1995), and interestingly, the osteoclasts have been shown to

resorb also carbonated hydroxyapatite free of pre-deposited proteins (Nakamura,

Hentunen, Salonen, Nagai, & Yamashita, 2013).

After attaching to the matrix and creating the ruffled border, the cells start

secreting hydrochloric acid (HCl) into the resorption area, as separate protons (H+)

and chloride ions (Cl-), which acidifies the lacuna (Cappariello et al., 2014). The

acidic environment dissolves the hydroxyapatite crystals leaving the bone’s organic

component exposed to be proteolytically processed (Vaananen, Zhao, Mulari, &

Halleen, 2000).

The organic bone matrix is degraded by lysosomal enzymes, which function in

the acidic conditions. The organic matrix degradation process is not completely

understood. The enzymes that have been suggested to be secreted into the

resorption lacunae are cathepsin K, TRAcP and multiple matrix metalloproteinases

(MMPs) (Henriksen et al., 2006; Rucci & Teti, 2016; Vaananen et al., 2000).

Cathepsin K is the primary cathepsin secreted by the osteoclasts and is thought to

be the main degrader of the bone’s main organic component the collagen type I

(Clarke, Drake, Oursler, & Khosla, 2017). Elevated cathepsin K levels are

associated with increased osteoclast activity and bone destruction (Skoumal et al.,

2004). TRAcP is the main intracellular osteoclast marker and it has also been shown

to function as a marker of osteoclastic activity in serum (Andersson et al., 2003;

Halleen, Tiitinen, Ylipahkala, Fagerlund, & Vaananen, 2006; Janckila, Takahashi,

Sun, & Yam, 2001). It has been suggested that osteoclasts secrete TRAcP into the

resorption lacuna, where it is believed to affect at least OPN and BSP function by

dephosphorylating them, which may affect the osteoclast attachment to the bone

matrix and migration (Andersson et al., 2003). The specific functions of the

multiple MMPs secreted into the resorption lacunae are not completely understood.

In general, MMPs have been suggested to take part in processing of both the

collagenous and NCPs of the bone matrix, as well as directly regulating osteoclasts

and other bone cells (Paiva & Granjeiro, 2017; Rucci & Teti, 2016).

During the matrix degradation, the degradation products and some of the

degrading enzymes are endocytosed into the osteoclast. Inside the cell the

endocytotic vesicles further fuse with more TRAcP containing intracellular vesicles,

and the degradation process continues (Halleen et al., 2006). The vesicles are then

transported to the functional secretory domain at the opposite side of the cell, where

they are exocytosed into the surrounding extracellular environment (Soysa & Alles,

Page 31: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

29

2016; Vaananen et al., 2000). A diagram of a resorbing osteoclast is shown in figure

3.

Fig. 3. A schematic presentation of a bone resorbing osteoclast. Osteoclasts attach to

the bone matrix and form the sealing zone. A resorption pit is formed under the ruffled

border membrane. Osteoclasts acidify the resorption pit by secreting H+-ions, and

matrix degrading enzymes to degrade the bone matrix. The matrix degradation products

are endocytosed and trafficked transcellularly to the functional secretory domain at the

opposite side of the cell and excreted. Modified from Cappariello et al., 2014 and Soysa

& Alles, 2016.

The transcytosis of degradation products allows osteoclasts to migrate and resorb

huge amounts of matrix during a resorption cycle when compared to their actual

size. It has been suggested that osteoclast migration during resorption is controlled

by the calcium concentration inside the resorption lacuna; after a certain

concentration is reached the cell moves slightly forward and starts resorbing again

(Cappariello et al., 2014; Teitelbaum, 2007). During an osteoclast’s life span the

cycle is repeated multiple times, and the osteoclast creates a resorption trail behind

it, as shown in figure 4. After multiple resorption cycles, at the end of their lifespan,

the osteoclasts undergo natural apoptosis (Rucci & Teti, 2016; Soysa & Alles, 2016).

The factors affecting or causing the osteoclast apoptosis are not known. Anti-

osteoporotic drugs such as zoledronic acid have been shown to cause osteoclast

apoptosis (Tai et al., 2017).

Page 32: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

30

Fig. 4. Electron microscope image of a resorbing osteoclast on a bone slice. On bone

osteoclasts migrate during resorption and leave a resorption trail behind.

2.1.4 Bone remodeling

Bone remodeling is fundamental to the quality of bone and maintenance of the

body’s mineral homeostasis throughout human life, as old bone is constantly being

degraded by osteoclasts and new bone produced by osteoblasts. A disturbance in

the balance may lead to a metabolic skeletal disease. As in osteoporosis, a common

disease, in which the balance is shifted towards increased osteoclastic activity

(Hadhidakis & Androulakis, 2006).

Bone remodeling is described to happen in cycles, which consist of five parts:

activation, resorption, reversal, formation and termination (Katsimbri, 2017;

Kenkre & Bassett, 2018). The cycles happen in an anatomical unit called the Basic

Multicellular Unit (BMU), which is composed of osteoclasts, osteoblasts, lining

cells and a capillary blood supply (Katsimbri, 2017; Kenkre & Bassett, 2018).

The bone remodeling activation and resorption parts consist of osteoclast

precursor migration to the BMU, osteoclastogenesis, osteoclast activation, and

bone resorption, with the local factors affecting them, as described earlier in chapter

2.1.3. The activation and reversal phases are also controlled by endocrine

hormones which control the body’s calcium and phosphate metabolism, such as

parathyroid hormone (PTH) and vitamin D. PTH and the active vitamin D

Page 33: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

31

(1α,25(OH)2D3) induce RANKL production in cells of the osteoblast lineage, thus

resulting in increased osteoclastogenesis (Silva & Bilezikian, 2015; Takahashi,

Udagawa, & Suda, 2014). Another important hormone is estrogen, which induces

apoptosis in the osteoclasts and is anti-apoptotic in the osteoblasts (Krum et al.,

2008). Estrogen promotes overall bone formation, and it has been shown that

estrogen deficiency in post-menopause is the major cause of osteoporosis in women

(Krum et al., 2008). Other important endocrine factors associated with bone

metabolism are glucocorticoids, calcitonin, growth hormone, thyroid hormone and

androgens (Kenkre & Bassett, 2018).

During the reversal phase, the pre-osteoblasts arrive at the location of resorbed

bone and differentiate into the osteoblasts that start producing new bone. This phase

of bone remodeling is not as well understood as the others (Katsimbri, 2017;

Kenkre & Bassett, 2018). It is likely that the before their apoptosis the osteoclasts

produce cytokines and other factors, the so-called clastokines, which attract cells

of the osteoblast lineage into the area to prepare the bone surface for new bone

formation by the mature osteoblasts (Cappariello et al., 2014). The first arriving

cells, called reversal cells, are of the osteoblast lineage and can further mature into

osteoblasts. However, they also have important functions of their own. The reversal

cells have been suggested to remove unmineralized collagen matrix, and then

deposit non-collagenous mineralized matrix, the “cement line”, in its place to

enhance the attachment and activity of the mature osteoblasts (Delaisse, 2014).

Unfortunately, the nature and function of the clastokines that affect cells of the

osteoblast lineage are not very well understood, as it has been apparent that even

though many cytokines produced by the osteoclasts increase osteoblastogenesis

and activity, many others decrease it, as explained earlier in 2.1.3 (Matsuo & Irie,

2008; Teti, 2013). Further research on this subject is needed.

After osteoblasts have arrived and matured at the BMU they begin the bone

formation phase. During this phase the removed old bone is completely replaced

with new bone. A disturbance in the osteoblast function at this point may lead to

decreased bone volume or density, e.g., post-menopausal estrogen deficient

osteoporosis. After enough new bone is formed, the final bone remodeling phase

termination is reached. During termination, the osteoblasts either undergo apoptosis,

become embedded in the matrix and differentiate into the osteocytes, or become

quiescent bone lining cells depending on their location and other bone cells present,

as described earlier in 2.1.2 (Katsimbri, 2017; Kenkre & Bassett, 2018).

Page 34: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

32

2.2 Joint

A joint is a site where two bones come together. In the human body there are two

kinds of joints, solid joints and synovial joints. In a solid joint two bones are

attached to each other by fibrous connective tissue or cartilage (Gray, 1918). The

movement in a solid joint is restricted. Such joints can be found, for example, in

skull sutures or at the pubic symphysis.

A synovial joint is a connection between two moving bones, which are

separated by an articular cavity, for example, a knee or a hip (Gray, 1918). The

articular cavity is formed by a joint capsule (Iwanaga, Shikichi, Kitamura, Yanase,

& Nozawa-Inoue, 2000). Usually the ends of bone are covered by articular cartilage,

and the joint capsule attaches to the margin of bone and cartilage (Iwanaga et al.,

2000). Other structures that can be found within the synovial capsule include

articular discs, fat pads, ligaments etc. (Gray, 1918). The shape of the synovial joint

varies according to its function from ball joints, which allow movement in all

directions, to plane joints, which allow only small sliding of the bones (Gray, 1918).

The articular cartilage which covers the end of the bones within the joint is

hyaline cartilage (Bhosale & Richardson, 2008). The cartilage, which is lubricated

by synovial fluid, creates a sliding surface between the opposing bones, (Bhosale

& Richardson, 2008). The hyaline cartilage is composed of chondrocytes, mostly

collagen type II fibers (90-95%) and other extracellular matrix proteins (5-10%)

(Kierszenbaum & Tres, 2015). Sixty-five to eighty percent of cartilage weight is

water (Bhosale & Richardson, 2008).

Chondrocytes are the main cartilage cells. They are responsible for the

production of the extracellular matrix (Kierszenbaum & Tres, 2015). The

chondrocytes are trapped within the cartilage, and because the hyaline cartilage is

avascular, they receive nutrients only by diffusion through the extracellular matrix

from the bone and the synovial fluid (Kierszenbaum & Tres, 2015).

The other extracellular matrix proteins, than collagens, are mainly

proteoglycans. They maintain the fluid and electrolyte balance within the cartilage

(Bhosale & Richardson, 2008). The proteoglycans can be divided into two: large

aggrecans and small proteoglycans, such as decorin, biglycan and fibromodulin

(Bhosale & Richardson, 2008).

The articular cartilage can be divided structurally and by chondrocyte

morphology into four layers from bone to articular cavity: calcified, deep, middle

and superficial zones (He et al., 2014). The extracellular matrix of the calcified

Page 35: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

33

cartilage is rich in calcium, and it acts as a transition zone between the bone and

the cartilage (He et al., 2014).

The joint capsule is formed by an inner synovial membrane and outer fibrous

membrane. The inner synovial membrane consists only of one to two layers of

synovial cavity lining cells on top of loose connective tissue, which is highly

vascularized and rich in lymphatic vessels (He et al., 2014). Further away from the

joint cavity the loose connective tissue merges into the denser connective tissue of

the outer synovial membrane (He et al., 2014). There are two types of synovial

lining cells: type A macrophage-like synovial cells and type B fibroblast-like

synovial cells (Iwanaga et al., 2000). The synovial cells do not have a continuous

base membrane or attach to each other tightly (Iwanaga et al., 2000). Normally, the

macrophage-like cells are in the minority, but in inflammation they can account for

up to 80% of the lining cells (Smith, 2011). The origin of the synovial macrophages

is suggested to be from the myeloid-monocyte line of cells, and the origin of the

fibroblast-like cells is thought to be mesenchymal (Kurowska-Stolarska &

Alivernini, 2017; Smith, 2011).

The synovial cells control the volume of the synovial fluid through the

production of synovial fluid glycoproteins and hyaluronic acid (Smith, 2011).

Synovial fluid is a viscous ultrafiltrate of plasma and proteins produced by the

synovial lining cells and the chondrocytes (Bennike et al., 2014). The main function

of synovial fluid is to work as a lubricant for the sliding articular surfaces, but it

also facilitates the transportation of nutrients and waste products between the

synovial lining and cartilage (Bennike et al., 2014).

2.3 Osteoimmunology

The interaction between the bone and the immune system was first discovered in

1972, when it was discovered that activated peripheral blood leukocytes release

agents that stimulate osteoclastic bone resorption (Horton, Raisz, Simmons,

Oppenheim, & Mergenhagen, 1972). Later multiple factors that regulate both the

bone and the immune cells have been found. Probably the most notable revelation

to date has been the discovery of RANK and RANKL, which were first discovered

as mediators of T-cell activation and dendritic cell function, but later were found to

be the most important regulators of osteoclastogenesis and osteoclast function

(Kong et al., 1999). The term osteoimmunology was first presented in 2000 to name

the field of study that investigates the connections between the bone and the

immune system (Arron & Choi, 2000). During this millennium osteoimmunology

Page 36: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

34

has been a rapidly growing and progressing field of study, which has resulted in

many great advances that have even led to production or on-going research of new

drugs, such as denosumab, an OPG analog, for treating osteoporosis, and multiple

biological RA drugs, for example, the TNF-α antibody adalimumab.

As the immune and the bone cells originate both from the same precursor cells

off bone marrow it is no wonder that complex signaling occurs between them. So

far, in inflammation the major interacting cells have been identified to be the

osteoclasts and the T-lymphocytes (Terashima & Takayanagi, 2018). During

inflammation the T-cells, and other inflammatory cells, activate and produce

multiple cytokines that either directly affect osteoclastogenesis or cause other cells,

such as fibrocytes or macrophages, to produce RANKL or other pro-osteoclastic

substrates, thus increasing the osteoclastogenesis (Okamoto & Takayanagi, 2019).

Cytokines that affect the osteoclastogenesis were discussed earlier at chapter 2.1.3.

The cytokines present in inflammation reflect its etiology and differ in different

situations, such as in OA or RA, and hence the effect on osteoclasts is likely to

differ between the diseases.

The osteoclastogenesis resulting from the increased inflammatory stimulus on

the pre-osteoclasts is called inflammatory osteoclastogenesis, which can especially

be seen in inflammatory joint diseases, such as RA, but is has also been suggested

to be behind secondary osteoporosis in chronic inflammatory diseases (Clayton &

Hochberg, 2013; Greisen et al., 2015; Suzuki et al., 2001). Interestingly the

inflammatory osteoclasts have been described to present a more macrophage-like

phenotype than the regular osteoclasts, as they are smaller and contain fewer nuclei

(Suzuki et al., 2001; Tsuboi et al., 2003).

The osteoclasts can also reciprocally affect the T-cells, but the total effect is

not fully understood and needs further research. Some studies have shown anti-

inflammatory regulation of T-cells by osteoclasts in both inflammation and cancer

(An et al., 2016; H. Li et al., 2014), but also reports of the osteoclasts further

activating the T-cells have been published (Pappalardo & Thompson, 2015). The

phenomenon is likely as complex as the combined effect of various cytokines either

increasing or decreasing the osteoclastogenesis.

The bone cells are crucial for the differentiation of the blood cells from

hematopoietic stem cells (HSCs) of the bone marrow. Osteoclasts provide the space

for the bone marrow and hematopoiesis within the bone by keeping the bone

remodeling in balance with the osteoblasts. It has been shown that osteopetrotic

mice suffer from high levels of extramedullary hematopoiesis due to lack of bone

marrow within the bones, and osteopetrosis is also associated with increased risk

Page 37: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

35

of inflammation and anemia (Lowell, Niwa, Soriano, & Varmus, 1996; Terashima

& Takayanagi, 2018). The osteoblasts also participate in HSC maturation and

differentiation by affecting the HSC homing, mobilization and number by

providing a bone platform to maintain them and by secreting different growth

factors that regulate the process (Kode et al., 2014; Okamoto & Takayanagi, 2019).

At this moment, advances in immune, osteoimmune and disease biomarker

research have led to discoveries of numerous proteins that may affect both the bone

cells and the immune cells, but their clinical significance in disease is uncertain,

whether they are actually an active player or an innocent by-stander

(Krishnamurthy et al., 2016; Lotz et al., 2013; J. W. Seol, Lee, Kim, & Park, 2009;

Yamamoto et al., 2003). In this study, we focus on the effects of the proteins OPN

and TRAcP, and their post-translational modifications, in inflammation and bone.

2.4 Osteopontin (OPN)

OPN is a phosphorylated N- and O-glycosylated non-collagenous extracellular

matrix protein (Christensen, Nielsen, Haselmann, Petersen, & Sorensen, 2005). It

belongs to the SIBLING protein family (Qin et al., 2004). Originally OPN was first

identified as a bone matrix protein in 1985 (Franzen & Heinegård, 1985), but later

it has been detected in almost all human tissues, and it is expressed by a large

variety of cells including all the bone cells (osteoblasts, osteocytes and osteoclasts),

macrophages, dendritic cells, chondrocytes, fibroblasts, endothelial cells, and

smooth muscle cells (Icer & Gezmen-Karadag, 2018; Sodek, Ganss, & McKee,

2000).

OPN regulates various cellular functions, e.g., activation, differentiation,

migration and adhesion throughout the human body, and plays a part in

inflammation, biomineralization, cardiovascular diseases, cancer, diabetes and

renal stone disease through different mechanisms (Brown, 2012; Ge et al., 2017;

Icer & Gezmen-Karadag, 2018; L. Wang et al., 2008). The known major biological

roles of OPN are explained in figure 5. OPN’s biological activity is controlled by

its post-translational modifications, especially phosphorylation, which is crucial for

most of its functions, such as immune cell or osteoclast activation (Addison, Masica,

Gray, & McKee, 2010; Kazanecki, Uzwiak, & Denhardt, 2007; Nyström, Dunér, &

Hultgårdh-Nilsson, 2007; Weber et al., 2002). OPN’s phosphorylation is controlled

extracellularly by TRAcP (Andersson et al., 2003).

Page 38: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

36

Fig. 5. The known major biological roles of OPN. Modified from Icer & Gezmen-Karadag,

2018.

The OPN protein consists of 300 amino acids, with some N- and O-linked

oligosaccharides. The protein contains an RGD sequence, two heparin binding

domains, one thrombin binding and one calcium binding domain (Kariya et al.,

2014; Christensen et al., 2005; H. Li et al., 2015). It has a total of 36

phosphoresidues; 34 phospho-serine and two phospho-threonine groups

(Christensen et al., 2005). OPN is secreted extracellularly in a phosphorylated form

after being phosphorylated intracellularly for example by Fam 20C kinase

(Christensen et al., 2005). It is known to have signaling functions through various

cell surface integrins, such as αvβ3 which can bind OPN’s RGD sequence, and a

CD 44 receptor (Kariya et al., 2014; Christensen et al., 2005; H. Li et al., 2015).

OPN can be cleaved into two active parts by thrombin, the N- and C-terminal ends,

which abilities are also dependent on the extent of their phosphorylation (Weber et

al., 2002). The C-terminal fragment contains the CD 44 receptor binding part and

it induces macrophage chemotaxis, while the N-terminal fragment binds integrins

and affects other cell spreading and activation (Weber et al., 2002).

In bone, OPN is known to affect osteoclast differentiation, activation and

migration (Ek-Rylander & Andersson, 2010; Ross et al., 1993; Sodek et al., 2000).

OPN is suggested to have an essential role in the adhesion of the osteoclasts to the

bone matrix and the generation of the ruffled border zone during bone resorption.

It has been shown that OPN can bind to bone hydroxyapatite (Goldberg, Warner,

Page 39: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

37

Li, & Hunter, 2001). The osteoclasts are believed to attach themselves to the

unresorbed bone through the αvβ3-integrin receptor and it is thought that the RGD-

sequence containing OPN is one of the key ligands for the αvβ3 in the bone matrix

(Ek-Rylander & Andersson, 2010; Ross et al., 1993; Sodek et al., 2000). It has been

shown that tge osteoclasts themselves can produce OPN, and it has been suggested

that the osteoclasts could facilitate their own attachment to hydroxyapatite by

producing OPN to function as a connecting molecule (Dodds et al., 1995; Maeda,

Kukita, Akamine, Kukita, & Iijima, 1994).

Osteoblasts are also capable of OPN production, but OPN’s extracellular

effects on osteoblasts are not well understood (Icer & Gezmen-Karadag, 2018).

OPN is likely to play an important role in osteoclast-osteoblast communication, as

the mineral content and crystallinity of bone in OPN knockout mice is increased

(Boskey, Spevak, Paschalis, Doty, & McKee, 2002). The mineral deposition by

OPN knockout osteoblasts has been shown to increased, but it can be decreased by

addition of extracellular phosphorylated OPN (Holm et al., 2014). Another study

showed that phosphorylated OPN might have suppressive effects on osteoblasts

that are under mechanical stress (Kusuyama et al., 2017). However, a study using

an osteoblast-like cell line (SaOS-2 cells) suggests that the suppressive effects of

OPN are also likely regulated by OPN’s phosphorylation. Dephosphorylated OPN

did not have the same effect as phosphorylated OPN on the SaOS-2 cells (Halling

Linder et al., 2017).

Along with OPN’s cellular functions, it also regulates biomineralization in its

free extracellular form (Gericke et al., 2005; Hoac et al., 2017; L. Wang et al., 2008).

The regulation happens in bone, but also in other sites involving calcification, such

as aortic valve calcification or urinary stone formation (Gericke et al., 2005; Hoac,

Nelea, Jiang, Kaartinen, & McKee, 2017; Kohri et al., 2012; Sainger et al., 2012).

Also, OPN’s effect on biomineralization is regulated by its phosphorylation, which

is required for its biomineralization inhibitory effect (Gericke et al., 2005; Hoac et

al., 2017; L. Wang et al., 2008). During mineralization OPN can effectively bind

ionic and crystalline calcium, and thus inhibit it. The key properties behind this

function are the protein’s large number of phosphorylation sites, highly acidic

nature, and intrinsically disordered structure (Hoac et al., 2017).

OPN’s expression is elevated during inflammation. It works as an important

chemoattractant especially for macrophages, neutrophils, dendritic cells, and T

cells (K. X. Wang & Denhardt, 2008). During inflammation OPN production is

increased in macrophages, activated T cells, epithelial and endothelial cells, and

smooth muscle cells (Icer & Gezmen-Karadag, 2018). Studies with OPN knockout

Page 40: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

38

mice have shown that OPN’s main immunoregulatory function is to regulate T-cell

activation and drive it towards a Th1 (T helper cell 1) cell-mediated response

instead of a Th2 (T helper cell 2) humoral response (Ashkar et al., 2000; K. X.

Wang & Denhardt, 2008). Th1 type T-cells produce the proinflammatory responses

responsible for killing intracellular microbes and for perpetuating autoimmune

responses. They regulate especially macrophage-monocyte function, as the Th2

responses balance these by promoting a humoral response which is more targeted

against extracellular parasites, by regulating eosinophils, basophils and mast cells

(Berger, 2000). Excessive Th1 T-cell activation is associated with increased tissue

damage during inflammation (Berger, 2000). OPN has also been shown to increase

Th17 cell differentiation and activity (G. Chen et al., 2010). Th17 cells differ from

Th1 and Th2 by predominantly producing IL-17 family cytokines, regulating

neutrophils, and they mainly target extracellular microbes and fungi (Ouyang,

Kolls, & Zheng, 2008).

It has been shown that in OPN null mice the severity of Herpes simplex,

Mycobacterium bovis BCG and Listeria monocytogenes-induced infections are

increased (Ashkar et al., 2000; Nau et al., 1999). Interestingly, OPN knockout mice

have a better survival from sepsis than normal mice (Fortis, Khadaroo, Haitsma, &

Zhang, 2015; Piao & Yuan, 2017). The increased survival is suggested to be a result

of an excessive inflammatory response in normal mice, and it has been suggested

that OPN’s protective effects in infection are overbalanced by its pro-inflammatory

capability. Similar results have been seen in inflammatory joint diseases. In RA

OPN levels decrease after one year of successful treatment (Izumi, Kaneko,

Hashizume, Yoshimoto, & Takeuchi, 2015). In addition, OPN deficiency has been

shown to reduce disease severity in arthritis mouse models (Yumoto et al., 2002).

In conclusion, research has shown that OPN plays an important role in many

biological functions and is an extremely versatile molecule present throughout the

human body. Especially, OPN has been of special interest to researchers working

on osteoimmunology, as it affects both bone and immune cells. However, due to

the extensive post-translational modifications and their effect on the protein’s

functions it has so far proven hard to completely understand the multiple functions

of OPN in the human body.

2.5 Tartrate-resistant acid phosphatase (TRAcP)

TRAcP is considered the main regulator of extracellular OPN’s phosphorylation

(Andersson et al., 2003), and thus the main regulator of OPN’s biological activity.

Page 41: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

39

A schematic presentation of how impaired TRAcP function in bone may lead to

pathological changes by affecting OPN’s phosphorylation is shown in figure 6. Two

forms of TRAcP can be found in the human body, TRAcP 5A and 5B. TRAcP is

synthesized as a relatively inactive monomer loop peptide pro-enzyme, TRAcP 5A.

The 5A form can proteolytically cleaved into a two-subunit active dimer form,

TRAcP 5B, by proteinases, such as cathepsin K, increasing its activity at least ten

times (Fagerlund et al., 2006; Ljusberg et al., 2005; Lång & Andersson, 2005). In

general, the TRAcP 5A form is thought to be produced by macrophages and

dendritic cells, and the 5B form by osteoclasts, thus 5A is the dominant form in

serum and 5B in bone (Halleen et al., 2006; Janckila & Yam, 2009).

TRAcP 5B is the main osteoclast marker and its levels in serum correlate with

osteoclast activity and number (Halleen et al., 2006; Janckila, Takahashi, Sun, &

Yam, 2001a). Elevated TRAcP 5B levels have been found in diseases associated

with increased bone metabolism and destruction, such as OA, RA, osteoporosis and

cancer with bone metastases (Chao, Wu, & Janckila, 2010; Halleen et al., 2002; J.

W. Seol et al., 2009; Tsuboi et al., 2003).

In bone TRAcP is suggested to control the osteoclast migration by regulating

their ability to attach to the bone matrix bound OPN by regulating OPN’s

phosphorylation status (Andersson et al., 2003; Ek-Rylander & Andersson, 2010).

By regulating OPN’s phosphorylation TRAcP also regulates the mineralization as

explained previously in chapter 2.3. TRAcP also partakes in the resorption and

degradation of bone matrix both in the resorption lacunae and intracellularly by

generating reactive oxygen species (Fagerlund et al., 2006; Andersson et al., 2003).

After secretion fro the osteoclasts TRAcP has been suggested to function also as a

clastokine which marks the newly resorbed resorption pit and acts as a docking

point for the osteoblasts and stimulates their activity (Cappariello et al., 2014;

Janckila & Yam, 2009).

TRAcP knockout mice exhibit a mildly osteopetrotic phenotype (Hayman et

al., 1996), as a likely result of weakened osteoclast activity. Transgenic mice

overexpressing TRAcP show increased bone turnover, but no change in bone

density or quality has been found (Hayman et al., 1996). The TRAcP inhibitors

developed have been suggested to possibly work as new osteoporosis drugs, but

are still to be clinically tested (Hussein, Feder, Schenk, Guddat, & P McGeary,

2018).

Page 42: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

40

Fig. 6. TRAcP regulates extracellular OPN’s biological activity by dephosphorylating it.

Impaired TRAcP activity may lead to pathological changes due to hyperphosphorylation

of OPN. In bone and related tissues phosphorylated OPN increases the activity of

immune cells and osteoclasts, decreases the activity of osteoblasts, inhibits

biomineralization, and increases the production of cartilage degrading enzymes in

chondrocytes. These pathological changes are seen in many bone related diseases,

such as RA, OA or osteoporosis.

TRAcP 5A’s clinical significance is not as well understood as 5B, but as it accounts

for the major part of TRAcP in serum it can be assumed to be effective, even though

it is not as potent an enzyme as the 5B form. 5A levels have been shown to correlate

with inflammation markers, such as CRP, ferritin, and triglycerides, and as the 5A

form is produced by macrophages it is suggested to be a special marker of their

activity (Janckila, Lederer, Price, & Yam, 2009). In recent research TRAcP 5A

levels have been correlated with chronic inflammation seen in cancer or in

metabolic syndrome (Y. Chen et al., 2015; Huang et al., 2017).

TRAcP is not only a marker of inflammation, but an active pro-inflammatory

regulator of cell function. However, TRAcP’s effects on inflammatory cells are not

as well understood as OPN’s. TRAcP null mice have been shown to have delayed

macrophage response to Staphylococcus aureus (Bune, Hayman, J Evans, & Cox,

2001) and impaired maturation of dendritic cells, which leads to deficient antigen

promotion, and thus impaired Th1 responses in T-cells (Esfandiari et al., 2006).

Page 43: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

41

In conclusion, TRAcP is the main regulator of OPN phosphorylation and its

most notable effects are mediated through its effects on OPN’s phosphorylation.

However, TRAcP also has independent functions in bone and inflammation that are

not well understood yet, and more studies are needed on both the 5A and 5B forms

of the enzyme. Further studies of TRAcP may reveal even a new therapeutic

prospective.

2.6 Rheumatoid arthritis (RA)

Rheumatoid arthritis is a chronic autoimmune inflammatory disease that affects

synovial joints. The characteristic pathological changes seen in RA are synovial

tissue inflammation, cartilage degradation, subchondral bone erosions, and

increased production of inflammatory cytokines (Weissmann, 2004). Typically, RA

is described as a symmetrical inflammation of small or intermediate synovial joints,

e.g., metacarpophalangeal joints in hands or wrist joints. It causes a substantial

burden on both the society and the patient. The patient’s burden results from

musculoskeletal deficits, and disease related comorbidities, such as cardiovascular

diseases or osteoporosis, which result in decreased quality of life and can be

debilitating (Smolen, Aletaha, & McInnes, 2016). The socioeconomic burden is a

result of direct medical costs, but also a consequence of functional disability

leading to decreased work and societal participation (Smolen et al., 2016).

The incidence and prevalence of RA varies between geographic areas and time.

In North America and Northern Europe the annual incidence rate is 20-50 per 100

000 inhabitants, while in Southern Europe the incidence is lower, 9.5-36 per

100 000 inhabitants (Scublinsky & Gonzalez, 2016). In Finland the incidence rate

is 41.6 per 100,000 inhabitants (Kononoff et al., 2017). The worldwide prevalence

of RA is considered to be just under 1%, with lower values in Mediterranean and

Scandinavian countries, 0.19-0.94 % (Scublinsky & Gonzalez, 2016). In Finland

the prevalence in the adult population is around 0.8% (Aho, Kaipiainen-Seppänen,

Heliövaara, & Klaukka, 1998). RA is two to three times more common in females

than men, and after pregnancy the incidence is increased by 2.3 times for the next

two years (Wallenius et al., 2010).

The etiology of RA is unclear, but it is thought be affected by both genetic and

environmental factors. The heritability of RA is estimated to be around 60%, of

which the HLA-antigens contribute 11-37% (Kurko et al., 2013). The

environmental factors suggested to play a part in RA are smoking, low economic

status, periodontitis, infectious agents such as Proteus mirabilis, Escherichia coli

Page 44: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

42

and Epstein-Barr virus, and changes in the bronchial or gut microbiome (Smolen

et al., 2016).

RA patients can be divided into two subgroups, by the level of rheumatoid

factor (RF) or anti-citrullinated protein antibodies (ACPAs), seropositive and

seronegative (Aletaha et al., 2010). The seropositive group is positive for either of

the biomarkers (Aletaha et al., 2010). The seropositive group is considered a more

homogenous group, due to a more similar disease etiology between the patients and

more aggressive symptoms (Aletaha et al., 2010). RA is associated with increased

mortality, especially from cardiovascular diseases (Dadoun et al., 2013). However,

with modern treatments the mortality has decreased close to the normal level today

(Listing et al., 2015), but RA still seriously affects an individual’s quality of life.

The pathogenesis of RA is not completely understood. The combinatory effect

of the environmental and the hereditary factors leads to the onset of the disease

(McInnes & Schett, 2011). The biomarkers of RA, such as ACPAs, RFs or increased

inflammatory markers have been shown to be circulating in the blood stream years

before the onset of the disease, especially in seropositive disease (Deane, 2014).

This condition is called preclinical RA, which is thought to be a result of altered

post-transcriptional modifications of different proteins, such as citrullination,

which leads to the generation of RA biomarkers (Deane, 2014). Various cells

implicated in RA pathogenesis, such as monocytes and osteoclasts, express the

citrullinated peptides on their cell surfaces, which are targeted by ACPAs (Shim,

Stavre, & Gravallese, 2018). This is suggested to lead to a loss of innate (e.g.,

macrophages, dendritic cells and neutrophils) and adaptive immunity (e.g., B- and

T-lymphocytes) tolerance (Calabresi, Petrelli, Bonifacio, Puxeddu, & Alunno, 2018;

McInnes & Schett, 2011). Later, an unknown trigger, an infection or other

environmental factor, leads to an onset of inflammation in the synovial tissue

(Calabresi, Petrelli, Bonifacio, Puxeddu, & Alunno, 2018; McInnes & Schett, 2011).

The joint swelling in RA is a result of the synovial tissue inflammation after

immune activation, which is characterized by leukocyte infiltration into the

synovial tissue. The inflammatory cells, such as T-cells and macrophages, produce

inflammatory cytokines, such as TNF-α, ILs and granulocyte-macrophage colony

stimulating factor (Gm-CSF), which lead to activation of synovial fibroblasts and

enhanced inflammatory cytokine production, which eventually leads to an

activation of osteoclasts (McInnes & Schett, 2011; Smolen et al., 2016). The

activation of osteoclasts leads to cartilage and bone damage, which causes a release

of pro-inflammatory substances and damage-associated molecular patterns

(DAMPs) (Orlowsky & Kraus, 2015). These in turn stimulate the innate immune

Page 45: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

43

system and immune cells even more, further increasing the synovial tissue

inflammation, and thus the osteoclasts activity, creating a vicious circle of

inflammation (Orlowsky & Kraus, 2015). Also, the areas of inflammation are

hypoxic and acidic, which promotes osteoclastogenesis and bone resorption (Arnett,

2010; Yuan et al., 2016).

There are no absolute diagnostic criteria for RA. The diagnosis is done

according to the symptoms and classification criteria by the American College of

Rheumatology/European League Against Rheumatism (Aletaha et al., 2010). The

typical clinical symptoms of RA, which are used to diagnose RA are symmetrical

synovial joint inflammation, typically in small or intermediate size joints, morning

stiffness and soreness, increased inflammatory parameters, and elevated ACPA or

RF levels in seropositive RA (Aletaha et al., 2010). In progressed disease typical

bone erosions can be seen especially in smaller joints at the bone areas which are

in contact with the synovial tissue (Schett & Gravallese, 2012). The erosions can

be visualized with native x-ray imaging (Aletaha et al., 2010). In larger joints, such

as knees, the erosions are not as large in relation to smaller joints, but a rapid

progression of so-called secondary OA can be seen, as shown in figure 7. It is

related to inflammation and the mechanism is different than in normal OA.

Page 46: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

44

Fig. 7. Native X-ray images of an RA patient’s knees. The patient was diagnosed with

RA in 2005. From 2009 to 2014 there has been a rapid progression of secondary OA in

the patient’s right knee. Due to the severity of the symptoms the patient underwent a

total knee replacement operation of the right knee.

After diagnosis anti-rheumatic immunosuppressive treatment is started

immediately. The goal of the treatment is remission of all symptoms within three

to six months (Rheumatoid Arthritis. Current Care Guidelines, 2015). The first line

of treatment suggested according to the Finnish Current Care Guideline is a

combination of methotrexate, sulfasalazine, hydroxychloroquine, small dose oral

glucocorticoid and local glucocorticoid injections in the affected joints. After a

long-sustained remission is reached the medication may be decreased slowly step-

by-step. Other conventional synthetic RA drugs include leflunomide, cyclosporin,

azathioprine, cyclophosphamide, mycophenolate and gold injections (Rheumatoid

Arthritis. Current Care Guidelines, 2015). If a combination of different

conventional synthetic drugs is not enough, a biological drug may be used

(Rheumatoid Arthritis. Current Care Guidelines, 2015; Smolen et al., 2017). A

Page 47: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

45

biological drug can be combined with methotrexate for better effect (Smolen et al.,

2017). The biological drugs for RA have five mechanisms of action: TNF-α

inhibition, interleukin 6 or 1 receptor inhibition, T-cell co-stimulation blockade,

and B-cell depletion (Smolen et al., 2016). Use of all biological drugs significantly

increases the risk of serious infections (Smolen et al., 2017).

Operative treatments are considered if conservative treatments fail, and their

goal is to relieve pain and improve function (Nikiphorou, Konan, MacGregor,

Haddad, & Young, 2014). They mainly include synovectomies, arthrodesis and

joint replacement operations. Operative synovectomy is done when a joint becomes

so inflamed that its movement and function are impaired, and it does not respond

to conservative treatments and the cartilage is still relatively healthy (Rheumatoid

Arthritis. Current Care Guidelines, 2015; Trieb & Hofstaetter, 2009). If the

cartilage is destroyed and operative treatment is needed the choice is usually

between arthrodesis and joint replacement depending on the affected joint and the

patient’s personal needs (Rheumatoid Arthritis. Current Care Guidelines, 2015;

Trieb & Hofstaetter, 2009).

Extra-articular co-morbidities associated with RA include osteoporosis,

cardiovascular disease, interstitial lung disease, vasculitis, secondary amyloidosis

and lymphoma (Rheumatoid Arthritis. Current Care Guidelines, 2015; Smolen et

al., 2016). The co-morbidities are often believed to be a result of both the chronic

inflammation and adverse effects of the treatments (Clayton & Hochberg, 2013;

Smolen et al., 2016). The increased mortality seen in RA is due to the increased

risk of cardiovascular diseases (Chung et al., 2005). However, with successful

treatment the risk can be reduced as the risk caused by the chronic inflammation is

higher than that of the treatments (Chung et al., 2005). Also, the risk of secondary

osteoporosis can be reduced with effective treatment (Clayton & Hochberg, 2013).

2.6.1 Osteoimmunology in RA

In RA the balance of bone metabolism between catabolism and anabolism is

heavily shifted towards catabolism, which is seen in the form of inflammatory bone

erosions caused by increased osteoclast number and activity (Favero et al., 2015).

The inflammatory synovial tissue is heavily infiltrated by myeloid cells of the

monocyte-macrophage lineage, which can differentiate into osteoclasts under the

stimulation of M-CSF and RANKL (Favero et al., 2015). The inflamed synovial

tissue cells at the bone-synovium interface express RANKL (Pettit, Manning,

Walsh, Goldring, & Gravallese, 2006), and even spontaneous generation of

Page 48: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

46

osteoclasts in synovial tissue and synovial monocyte cultures in vitro has been

reported (Greisen et al., 2015; Suzuki et al., 2001).

RANKL knockout mice are protected from bone erosions in inflammatory

arthritis models (Pettit et al., 2001), and it is a general consensus that RANKL is

the main factor behind the inflammatory osteoclastogenesis and increased

osteoclastic activity in RA (Favero et al., 2015; Okamoto & Takayanagi, 2019;

Shim et al., 2018). Other inflammatory factors, such as inflammatory cytokines and

proteins, affect osteoclast differentiation and activity either by driving the

inflammatory or synovial cells to produce RANKL, by affecting RANK/RANKL

signaling in the pre-osteoclasts or by direct cellular mechanisms (Favero et al.,

2015; Okamoto & Takayanagi, 2019; Shim et al., 2018). An example of factors that

can directly affect osteoclasts are ACPAs, which can directly bind into osteoclasts

or their precursors and increase their differentiation and activity, and high blood

ACPA levels correlate with aggressive disease phenotype and bone erosions (Harre

et al., 2015).

Most of the important cytokines increased in RA are also associated with

increased osteoclast activity, such as TNF-α, IL-6, IL-8, IL-17, VEGF etc.

(Amarasekara et al., 2018; Barra, Summers, Bell, & Cairns, 2014; Hampel,

Sesselmann, Iserovich, Sel, Paulsen, & Sack, 2013). Also increased pro-

inflammatory cytokine levels have been shown to correlate with the disease

severity (Barra et al., 2014; Hampel, Sesselmann, Iserovich, Sel, Paulsen, & Sack,

2013). However, as in all inflammation, there is also an active anti-inflammatory

component in RA, and many cytokines shown to be increased in RA are also

associated with decreased osteoclast function, such as IFN-α, IFN-γ, IL-3, Il-10,

IL-27 and IL-33 (Amarasekara et al., 2018; Khan, 2018; Lai et al., 2016; Pavlovic,

Dimic, Milenkovic, & Krtinic, 2016; Sellam et al., 2016; Zhao, Huang, & Zhang,

2016). Overall, the pro-osteoclastic stimulus is stronger, as can be seen in the

clinical bone erosions. However, because the RANKL effect on osteoclasts is so

much stronger than the other factors, it has been hard to evaluate the combined

effect of the other factors, even though the RA drugs target factors other than

RANKL.

In our study OPN is of special interest. OPN’s levels have been shown to be

elevated in serum, synovial fluid and urine of RA patients (Hasegawa et al., 2009;

Iwadate et al., 2013; Shio et al., 2010). Serum OPN levels have also been shown to

correlate with bone resorption markers, such as TRAcP 5B and C-terminal

telopeptide (CTX), and as such to correlate with disease severity (Iwadate et al.,

2013). OPN expression has been indicated in RA synovial tissue (Hasegawa et al.,

Page 49: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

47

2009). OPN knockout mice suffering from inflammatory arthritis, were shown to

be protected from articular cartilage and inflammatory bone loss (Yumoto et al.,

2002). Overall, OPN plays a crucial role in the inflammatory joint diseases, such

as RA, but its clinical significance is not well understood yet.

In synovial tissue, overexpression of OPN during inflammation, especially in

synovial fibroblasts and T-cells, leads to further inflammatory cell activation, which

leads to elevated production of pro-inflammatory cytokines, including OPN itself

(G. Xu et al., 2005). In RA OPN is shown to upregulate Th1 and Th 17 T-cell

activation and function (G. Chen et al., 2010). Both Th1 and Th17 activation and

the related cytokine production activation are associated with the autoinflammatory

pathogenesis and increased bone degradation in RA (Bazzazi et al., 2018). Other

inflammatory cells that OPN activity directly increases are macrophages,

neutrophils and dendritic cells (K. X. Wang & Denhardt, 2008). Also, OPN directly

increases osteoclast differentiation, migration and activation, as previously

discussed in chapter 2.4 (Ek-Rylander & Andersson, 2010; Ross et al., 1993; Sodek

et al., 2000).

TRAcP levels have been shown to be elevated in RA (Iwadate et al., 2013;

Janckila, Neustadt, & Yam, 2008). As previously discussed in chapter 2.4, OPN’s

biological activity is dependent on its phosphorylation, which is controlled by

TRAcP. Interestingly, even though both TRAcP and OPN levels increase in RA, the

effect of TRAcP on OPN’s phosphorylation, and, thus on its biological activity has

not been assessed. In this study, one of our goals was to evaluate the

phosphorylation of OPN in RA.

The therapeutic properties of anti-OPN treatments have been investigated and

are under research in RA animal models, and some results have been encouraging

(Zhang, Luo, Li, Gao, & Lei, 2015), but more studies are necessary for further

clarification of OPN’s role in RA and the development of OPN-targeted RA therapy.

2.7 Osteoarthritis (OA)

Osteoarthritis is the most common joint disease in the world. It affects 10% of men

and 18% of women over 60 years of age and causes a huge socioeconomical burden

(Woolf & Pfleger, 2003). In Finland the age adjusted prevalence of knee OA is 6.1%

in men and 8.0% in women over 30 (Arokoski et al., 2007). In the age group 75-

84, 15.6% of men and 32.1% of women have knee OA (Arokoski et al., 2007).

Clinical knee OA is rare in people under 45 years, as in the age group 30-44 the

prevalence is only 0.3% in men and 0.4% in women (Arokoski et al., 2007).

Page 50: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

48

Traditionally OA has been considered to be a result of prolonged mechanical

wear on joint articular surfaces, which results in a loss of articular cartilage,

pathological changes in subchondral bone areas and reactive synovial tissue

inflammation. However, advances in modern research have revealed that OA is

actually a very complex disease of the whole joint associated with genetic,

biological, and biomechanical components, and the etiological factors are joint

specific (Glyn-Jones et al., 2015). Most often OA affects weight bearing joints,

such as knees and hips, or smaller joints, such as finger joints that are extensively

used, but any synovial joint in the body can be affected. To date the fundamental

etiology of OA is unknown.

The main risk factor of OA is age, as the other primary factors that cause OA

must generally affect over a longer period of time. Factors that have been associated

with the risk of OA are female gender after menopause, obesity, genetics, diet,

trauma, abnormal or extensive loading of joints and structural abnormalities

(Georgiev & Angelov, 2019; Palazzo, Nguyen, Lefevre-Colau, Rannou, &

Poiraudeau, 2016). Interestingly, obesity does not only increase the risk of OA in

weight bearing joints but also in hands, indicating that obesity has metabolic and

inflammatory systemic effects also on the joints (Palazzo et al., 2016). The genetic

factors have been suggested to account for 60% of hand and hip OA and 40% of

knee OA (Palazzo et al., 2016). Diseases that increase the risk of OA are generally

associated with metabolic syndrome, such as diabetes, cardiovascular diseases and

depression (Georgiev & Angelov, 2019). OA is not associated with increased risk

of osteoporosis (Clayton & Hochberg, 2013; Im & Kim, 2014).

In early OA, the synovial cells are triggered to produce inflammatory cytokines

and mediators, which activate the chondrocytes and make them to produce cartilage

degrading enzymes, such as MMPs, and more inflammatory cytokines, such as IL-

1β, IL-6 and TNF-α (Glyn-Jones et al., 2015; Sulzbacher, 2013). The inflammatory

mediators in cartilage can ultimately cause chondrocyte hypertrophy and apoptosis

(Sulzbacher, 2013). The cartilage degradation causes a release of pro-inflammatory

substances and DAMPs into the synovial fluid. This in turn stimulates the innate

immune system and immune cells in the synovial tissue, which further increases

the synovial tissue inflammation. The circle of inflammation in OA joint very much

like that seen in RA, but in OA the inflammation is generally significantly weaker

(Orlowsky & Kraus, 2015). Along with the cartilage degradation and the synovial

tissue inflammation, OA is characterized by changes in the subchondral bone

remodeling and resorption in the form of local subchondral bone sclerosis, and by

the formation of osteophytes and cysts. Osteophytes are exostoses that form along

Page 51: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

49

the joint margins. In OA, the production of RANKL and various other signals that

affect osteoclasts are also upregulated in synovial tissue and their levels increased

in synovial fluid (Hampel, Sesselmann, Iserovich, Sel, Paulsen, & Sack, 2013;

Pilichou et al., 2008). The changes in the subchondral bone start already in early

OA and can advance throughout the disease (Glyn-Jones et al., 2015; Sulzbacher,

2013). In advanced OA, the articular cartilage can be completely degraded and the

joint may become ankylosed (Kellgren & Lawrence, 1957).

The most characteristic symptom of OA is joint pain, which typically is worse

during exercise and eases at rest, but in advanced disease the pain can become

continuous even at night (Knee and Hip Osteoarthritis. Current Care Guidelines,

2018). Morning stiffness of the affected joints, especially in the hands is also typical

(Knee and Hip Osteoarthritis. Current Care Guidelines, 2018). In advanced disease

a joint’s range of motion can become decreased. The stronger the symptoms are the

more they limit the patient’s daily activities and severe OA symptoms can be

debilitating.

The typical bone changes in OA are increased cortical bone thickness,

deformation and flattening of the articular shape, decreased mass of subchondral

trabecular bone, bone marrow lesions, subchondral cysts, tidemark duplication and

osteophyte formation (Favero et al., 2015). Bone marrow lesions are subchondral

bone oedema, so-called bone bruises, which can be seen with magnetic resonance

imaging in early OA. (Alliston, Hernandez, Findlay, Felson, & Kennedy, 2018).

Tidemark duplication means the advancement of the calcified cartilage further

away from bone in OA (Alliston et al., 2018). The pathological bone changes in

OA are either osteolytic or sclerotic, which gives OA two types of phenotypes,

osteoporotic or osteosclerotic, that can both co-exist in the same patient even at

different parts of the same joint (Favero et al., 2015). Subchondral bone turnover is

increased 20-fold in OA (Bailey, Mansell, Sims, & Banse, 2004). The increased

subchondral turnover is a result of increased osteoclastic and osteoblastic activity,

which are thought to be results of increased mechanical stress and inflammation in

the joint (Hügle & Geurts, 2016). However, in OA the subchondral bone is

hypomineralized and of lesser quality compared to normal subchondral bone

(Bailey et al., 2004).

The diagnosis of OA is done from native x-ray images (Knee and Hip

Osteoarthritis. Current Care Guidelines, 2018). The radiologic findings in OA are

joint space narrowing, osteophytes, subchondral bone sclerosis and bone

deformities in advanced disease (Kellgren & Lawrence, 1957). The most common

radiological grading used for staging OA is the Kellgren-Lawrence grade, which

Page 52: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

50

grades the radiological severity of OA as grades I-IV according to the above

radiologic changes (Kellgren & Lawrence, 1957).

As the etiology of OA is unknown and the pathogenesis is not completely

understood, there are no specific curative or disease progression preventing

treatments available. The treatments are focused on managing the symptoms and

improving the patients’ quality of life. Conservative drug free treatments are the

basis of OA treatments. Lifestyle changes that promote healthy lifestyle, exercise

and weight loss are recommended for each patient (Glyn-Jones et al., 2015; Knee

and Hip Osteoarthritis. Current Care Guidelines, 2018). Physical or manipulative

therapy may be used to help symptoms (Knee and Hip Osteoarthritis. Current Care

Guidelines, 2018). Joint pain can be treated with paracetamol, non-steroidal anti-

inflammatory drugs, intra-articular corticosteroid or hyaluronate injections, and

opioids according to the severity of symptoms (Knee and Hip Osteoarthritis.

Current Care Guidelines, 2018). Surgical treatments, such as prosthesis operation,

osteotomies to correct a joint’s mechanical axis or osteosynthesis can be performed

in patients with progressed disease and difficult symptoms according to the

required standard of function and affected joint (Glyn-Jones et al., 2015; Knee and

Hip Osteoarthritis. Current Care Guidelines, 2018). Biological drugs, such as anti-

TNF-α, used for RA have been suggested and tested as new drugs for OA, but they

have shown only mild benefit and are not recommended to be used at present, while

further studies on biological drugs are conducted (Dimitroulas, Lambe, Klocke,

Kitas, & Duarte, 2017).

2.7.1 Osteoimmunology in OA

The synovial inflammation seen in OA is very much like that seen in RA or other

inflammatory arthritis (Dar, Azam, Anupam, K Mondal, & Srivastava, 2018; Hügle

& Geurts, 2016). In OA, as in RA, there is also an abundance of inflammatory

monocytes/macrophages in the synovial tissue (Mathiessen & Conaghan, 2017).

As earlier discussed, it is generally thought that some triggering event causes an

activation of the innate immunity in the synovial tissue, which leads to the

activation of the inflammatory cells (Orlowsky & Kraus, 2015). Various studies

have revealed that the inflammatory T-cell subtypes activated in OA resemble the

ones activated in RA, with elevated percentages of Th1, Th9 and Th17 and

decreased T-regulatory cells in OA patients as compared to healthy controls (Dar et

al., 2018). The activation of inflammatory cells and synovial cells due to

inflammation leads to the production of inflammatory cytokines and related

Page 53: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

51

proteins. The proinflammatory cytokines most importantly associated with OA are

IL-1β, TNF-α, IL-6, IL-15, IL-17, and IL-18 (Wojdasiewicz, Poniatowski, &

Szukiewicz, 2014). The production of RANKL in the synovial cells and its

concentration in synovial fluid is also increased in OA, when compared to healthy

controls (Monasterio et al., 2018; Pilichou et al., 2008). As in all inflammation, also

increases of anti-inflammatory cytokine levels, most notably IL-4, IL-10, and IL-

13, are seen in OA (Wojdasiewicz et al., 2014). Overall, the combined effect of

increases of inflammatory cytokines along with the increased RANKL levels

increase osteoclast differentiation and activity in a similar fashion as in RA.

However, the strength of the inflammation and the levels of inflammatory cytokines

in RA are generally significantly higher than in OA (Hampel, Sesselmann,

Iserovich, Sel, Paulsen, & Sack, 2013). Thus, their effect on bone cells is stronger

and the changes seen in the bone often happen faster in RA and are more clearly

seen at locations in contact with the synovial tissue, especially in smaller joints. In

OA, the bone turnover is increased in wider areas throughout the joint and the

clinical changes seen in bone generally develop over a longer time.

OPN levels have been shown to be elevated in both the synovial fluid and the

serum of OA patients when compared to healthy controls (Cheng, Gao, & Lei,

2014), and increased OPN levels have been shown to correlate with the severity of

joint lesions and inflammation in OA (Gao et al., 2010; Honsawek, Tanavalee,

Sakdinakiattikoon, Chayanupatkul, & Yuktanandana, 2009). The synovial

fibroblasts have been shown to produce OPN in OA (Zhang et al., 2013). The

increased OPN levels in the OA synovial tissue are likely to function in a similar

fashion as in RA, where they increase the inflammatory cell function, especially

Th1 and Th 17 cells (G. Chen et al., 2010). This would also result in the increases

of the cytokines associated with OA. OPN has also been suggested to directly

interact with the chondrocytes and cause a release of cartilage degrading MMPs

(Cheng et al., 2014). Interestingly, OPN deficient mice have been shown to be more

prone towards joint instability and aging related OA (Matsui et al., 2009), even

though OPN knockout mice suffering from inflammatory arthritis were shown to

be protected from the articular cartilage and the inflammatory bone loss (Yumoto

et al., 2002), which may indicate an inadequate response of bone and cartilage cells

towards mechanical stress.

The expression of OPN’s phosphatase, TRAcP, in the damaged chondrocytes

and concentration in the synovial fluid of OA patients has been shown to be

increased (Seol et al., 2009). The phosphorylation of OPN in synovial fluids of OA

patients has not been assessed. However, increased phosphorylation of OPN in the

Page 54: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

52

articular cartilage has been shown, when compared to healthy cartilage, and the

increased phosphorylation of OPN to lead to increased MMP production in the

chondrocytes (Xu et al., 2013).

Anti-OPN treatments for OA have been suggested, and they have been shown

to possibly decrease inflammation, cartilage degradation and bone changes (Cheng

et al., 2014), but elucidating the therapeutic aspect of OPN needs further studies

and better knowledge of OPN’s role in the pathogenesis of OA.

Page 55: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

53

3 Aims of the study

Osteoimmunology is a field of study which focuses on the connection of the bone

and the human immune system. Rheumatoid arthritis and osteoarthritis are

common joint diseases which cause enormous amounts of human suffering and

huge economical burdens throughout the world. Their pathogenesis is not fully

understood, and we are limited to treating the symptoms of the diseases as no

curative treatment is known. The etiology and pathogenesis of the diseases are

different, but both are associated with changes in bone metabolism and bone

destruction. To study how the diseases affect bone metabolism, one must know how

the bone cells function. Therefore, the basic functions of osteoclasts and different

proteins that affect them were studied, along with the effect of the studied diseases

on osteoclasts.

The specific aims of the study were to examine:

1. The phosphorylation of OPN in RA synovial fluid.

2. Secretion of OPN by human osteoclasts into the resorption pits on human bone

during resorption.

3. The effect of RA and OA patients’ synovial fluid and serum on

osteoclastogenesis and bone resorption in vitro.

Page 56: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

54

Page 57: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

55

4 Materials and methods

The methods used in this thesis are summarized in the table 1 below and described

in detail with references in the original articles I-III.

The patient samples for the studies were collected in Oulu University Hospital.

All the patients were informed of the study and gave a written approval for the use

of their samples. The healthy control samples were collected from healthy informed

volunteer donors. The protocol followed the Helsinki Declaration principles in full,

and the Northern Ostrobothnia Hospital District Ethical Committee provided an

approval for the study and tissue collection. The process of sample and material

acquisition for each individual study is described in detail in the original articles I-

III. The number of patient samples for each study are listed in the results. Other

materials used are described in the original articles.

For study I, synovial tissue biopsies and fluid were obtained from RA and OA

patients. The phosphorylation of OPN in synovial fluid was analyzed using the

western blot method and the concentrations of TRAcP 5A and 5B forms were

analyzed using an enzyme-linked immunosorbent assay (ELISA).

Immunohistochemistry and light microscopy were used to assess OPN and TRAcP

5A/5B forms in the synovial tissue.

In study II, human peripheral blood and bone marrow derived osteoclasts were

cultured on human bone and carbonated hydroxyapatite slices. Bone resorption and

OPN were quantified from the surface of the resorbed slices using field emission

scanning electron microscopy (FE-SEM) and gold labeling of OPN. A variety of

fluorescently labeled lectins were used to stain the glycan epitopes present in the

resorption pits. Mass spectrometry was used to analyze the proteomics of the

resorbed bone slices. The effect of enzymatic removal of α2,3 and α2,6 sialic acids

from the surface of pre-resorbed bone slices on osteoclasts was also analyzed.

Finally, immunohistochemistry of osteoarthritic femoral heads was done to analyze

the localization of OPN and TRAcP in sites of increased bone metabolism in vivo.

In study III, human peripheral blood monocyte derived osteoclasts, obtained

from a healthy donor, were cultured on bone slices with the synovial fluid samples

acquired from RA and OA patients undergoing knee surgery, and with serum from

untreated RA patients collected at the time of diagnosis and healthy volunteer

controls. Supraphysiological RANKL concentrations were used to evaluate the

effect of other inflammatory factors. The osteoclast differentiation and the bone

resorption were evaluated using light and laser microscopy. The concentrations of

27 different cytokines and related proteins were analyzed from the RA and OA

Page 58: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

56

patient synovial fluids, and the untreated RA patient and the healthy control serums

with a multiplex assay. The synovial fluid OPG and RANKL concentrations were

analyzed using a simplex assay.

Table 1. Methods. The materials used are explained in each study.

Level Method Used in

Protein Western blot I

ELISA (Enzyme-linked immunosorbent assay) I, III

Multicytokine assay III

Cells and tissues Cell culture II, III

Preparation and staining of tissue samples and cells I, II, III

Mass spectrometry II

Electron microscopy II

Laser microscopy III

Confocal microscopy II, III

Light microscopy I, II, III

Other Statistical analyses I, II, III

The statistical analyses for the articles were done using IBM SPSS Statistics

versions 20 and 24 (IBM, NY, USA). A specialist statistician was consulted about

the analyses. Depending on the distribution of the values, parametric or non-

parametric tests were used. In study I non-parametric Mann-Whitney U-test and

Spearman’s correlation were used due to the low number of study subjects and non-

normal distribution of results. In study II the results were normally distributed, and

student’s t-test was used. In study III nonparametric tests, Independent Samples

Median Test and Mann-Whitney U test, and Fisher’s exact test for nominals were

used due to the low number of samples. In all studies a p-value less than 0.05 was

considered statistically significant.

Page 59: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

57

5 Results

5.1 Analyses of OPN and TRAcP in RA synovial fluid and tissue (I)

Synovial fluid and tissue biopsies were acquired from RA and OA patients during

knee prosthesis operations. OPN’s phosphorylation level and the concentrations of

TRAcP 5A and 5B forms were analyzed from the synovial fluid, and

immunohistochemistry was conducted to show the presence of OPN and TRAcP in

synovial tissue. The hypothesis was that OPN is more phosphorylated in RA

synovial fluid, which leads to the stronger immune cell and osteoclast activation as

seen in RA when compared to OA. The samples analysed are shown in table 2.

Table 2. Patient samples analyzed in the study I.

Study Sample n

OPN in synovial fluid (Fig 1A, B and C) seropositive RA patient synovial fluid 9

seronegative RA patient synovial fluid 7

OA patient synovial fluid 5

TRAcP 5A/5B in synovial fluid (Fig 2D, E and F) seropositive RA patient synovial fluid 14

seronegative RA patient synovial fluid 10

OA patient synovial fluid 24

Synovial tissue immunohistochemistry (Fig 3) Representative RA biopsies 5

Representative OA biopsies 16

In study I tissue biopsies and synovial fluids were taken from the same patients, but

unfortunately many of the biopsies, especially the samples from RA patients, were

of poor quality and the number of representative biopsies that could be analyzed

was reduced. The difference in the number of samples analyzed for OPN and

TRAcP is due to the low volume and availability of samples at the time of analysis.

5.1.1 Phosphorylation of OPN in synovial fluid (I)

The western blotting method was used to analyze the level of OPN’s

phosphorylation in the synovial fluids of OA and RA patients. Full-length OPN was

significantly more phosphorylated in both seropositive and seronegative RA

patients’ synovial fluid, when compared to OA patients’ synovial fluid (Fig 1A in

I). The total levels of OPN did not differ between the patient groups (Fig 1B in I).

Page 60: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

58

We also showed that the thrombin cleaved C-terminal end of the OPN fragment

was similarly more phosphorylated in both RA groups when compared to OA (Fig

1C in I).

5.1.2 TRAcP 5A and 5B concentrations in synovial fluid (I)

To explain the differences seen in OPN’s phosphorylation ELISA was used to

measure the concentrations of TRAcP 5A (Fig 1D in I) and 5B (Fig 1E in I) forms

from the synovial fluid samples of RA and OA patients. The overall combined

amount of both TRAcP forms in the synovial fluids was the same between all

sample groups; seropositive and seronegative RA, and OA (Fig 1F in I). However,

in the seropositive RA samples the ratio of TRAcP 5B/5A forms was decreased

when compared to OA samples. Seropositive RA patients had more less active 5A

than active 5B form (mean TRAcP 5B/5A ratio 0.714±0.259). The OA synovial

fluids had more active 5B than less active 5A form (mean TRAcP 5B/5A ratio

1.384±0.275) (Fig 2A in I).

When comparing the samples in which the TRAcP 5B/5A ratio and the

phosphorylation of OPN were both measured, a significant negative correlation

between the phosphorylation of OPN and the TRAcP 5B/5A ratio was found (Fig

2B in I). A decreased TRAcP 5B/5A ratio led to increased phosphorylation of OPN

in RA synovial fluid.

5.1.3 Localization of OPN and TRAcP in RA and OA patient synovial

tissue (I)

OPN was localized mostly in the extracellular matrix in the synovial tissue samples.

The staining for OPN was the most intense in the sublining layer of synovial tissue,

with lighter staining present in the lining layer of the joint cavity and deeper stroma

(Table 2, Fig 3J and K in I). Areas containing large quantities of inflammatory cells,

such as lymphocytes and macrophages, also showed strong intracellular staining.

No differences in staining were found between RA and OA samples (Table 2 in I).

Visualization of TRAcP in synovial tissue was done at the same time with two

different antibodies, one binding both 5A and 5B forms (Fig 3A, B, E and F in I)

and the other capable of binding specifically the TRAcP 5A form (Fig 3C, D, G and

H in I). Staining for both antibodies was strongest at the synovial lining layer and

weakest at the sublining layer. Average staining was present also in the deeper

stroma. The staining of the sublining layer was slightly stronger with the TRAcP

Page 61: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

59

antibody binding both forms in OA than in RA samples: no other differences were

found between RA and OA samples (Table 2 in I). This small difference may

indicate that there is more TRAcP 5B in the sublining layer of OA synovial tissue

when compared to RA, which would be in accord with the measured TRAcP 5B/5A

ratio differences between OA and RA synovial fluid samples. However, this is only

speculative based on the histological samples, and no definite conclusions can be

drawn. No unspecific staining was seen in control samples, for which the staining

protocol was done without the primary antibodies, as shown in figure 8.

Fig. 8. Synovial tissue section staining without primary antibody. No unspecific

staining was seen.

An EnVision Detection Systems Peroxidase/DAB, Rabbit/Mouse, HRP.

Rabbit/Mouse (DAB+) kit was used according to the manufacturer’s instructions

for the stainings, and the samples were studied visually under a light microscope.

Page 62: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

60

5.2 Analyses of OPN in resorption lacunae (II)

The effects of extracellular OPN on osteoclasts are far better known than those of

endogenous osteoclast OPN. Earlier studies have suggested that osteoclasts deposit

OPN into the resorption pit during bone resorption to facilitate their attachment into

the resorbed matrix, as explained in chapter 2.3. However, no previous studies with

human derived cells on human bone have been conducted to investigate OPN’s

deposition by osteoclasts.

In study II, human peripheral blood and bone marrow derived osteoclasts were

cultured on human bone and carbonated hydroxyapatite slices to study the

deposition of OPN by the osteoclasts during resorption. Carbonated hydroxyapatite

is a material that osteoclasts can resorb, but which is free of pre-deposited proteins

to avoid confounding by revelation of pre-deposited bone osteopontin during bone

resorption.

The glycan epitope at the bottom of the resorption pit was also studied to reveal

other signals present after resorption. Proteomics analysis of the resorbed bone

slices was conducted to distinguish and measure the proteins increased in the

resorbed bone.

Table 3. Samples analysed in study II.

Study Sample n

FE-SEM detection of OPN in resorption pit Resorbed bone slices 4

(Fig 1 in II) Resorbed carbonated hydroxyapatite slices

Total number of resorbed slices

2

6

Proteomics analysis of resorbed bone slices Resorbed bone slices 2

(Fig 3 in II) Number of mass spectrometry runs per slice 6

Lectin stainings of resorbed bone slices Resorbed bone slices per lectin 3

(Fig 2 a-l in II)

Osteoclast differentiation on enzymatically treated

human bone slices (3 cultures from different bone

marrow donors) (Fig 2 m-n in II)

Total number of resorbed bone slices

Resorbed bone slices

Un-resorbed bone slices

21

9+6+4

9+6+4

5.2.1 Detection of OPN in resorption lacunae using FE-SEM (II)

FE-SEM and immunodetection of OPN with gold particles were used to study the

deposition of OPN into the resorption pits on human bone and carbonated

hydroxyapatite by human osteoclasts. Actively resorbing osteoclasts (Fig 1a in II),

both bone marrow and peripheral blood monocyte derived, created multiple

Page 63: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

61

resorption lacunae on both the bone (Fig 1c and e in II) and the hydroxyapatite (Fig

1b and d). OPN was significantly enriched in the resorption pits on both resorbed

substrates indicating that osteoclasts deposit OPN into the resorption lacunae

during resorption (Fig 1e, d and f in II). On bone, the number of OPN particles in

the electron microscope view with 20k magnification inside the pit was increased

2.0-3.7 times and on carbonated hydroxyapatite 2.8-12 times. The real difference

is likely larger as the bottom of the resorption pit is not even and the area on which

the electron microscope can focus is reduced as seen in Fig 1d and e in II. Due to

the low number of samples that it was possible to produce for analysis at a single

time, the study was repeated with similar results.

5.2.2 Analysis of proteins enriched in the resorbed bone slices (II)

Proteomics analysis of the resorbed bone was conducted using mass spectrometry

to distinguish the proteins that are enriched in the resorbed bone, and to measure

how much the proteins are enriched. The cells were mechanically removed from

the bone slice before the analysis. Un-resorbed bone slices were used as controls.

Multiple proteins where found enriched, of which OPN was the most enriched

(Fig 3b in II). Along with OPN, the proteins that were notably enriched in the

analyzed bone slice samples were prolargin, asporin and mimecan. OPN was

increased ten fold, prolargin and asporin seven fold, and mimecan four fold. OPN’s

phosphorylation was not studied, and it may be possible that heavily

phosphorylated forms of OPN may not have been detected.

5.2.3 Analysis of glycan epitope in the resorption lacunae (II)

The glycan epitope at the bottom of resorption pits on human bone resorbed by

human bone marrow and peripheral blood derived osteoclasts in vitro was studied

using different test plant lectins and light microscopy (Fig 2a-l in II). The cells were

mechanically removed from the bone slices before the analyses. An abundance of

α2,3 and α2,6 -linked sialic acids were found at the bottom of resorption lacunae.

The sialic acid residues were the highest in O-glycans. OPN is an O-glycosylated

α2,3 and α2,6 -sialic acids containing protein, and being the most enriched protein

in resorbed bone slices, it is most likely the protein presenting the sialic acids the

lectins bind.

When the α2,3 and α2,6 sialic acids were enzymatically removed from the

resorbed bone slice surface and osteoclasts re-cultured on them, the number of

Page 64: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

62

osteoclasts was doubled, when compared to cultures where the sialic acids were

still present on the surface of the pre-resorbed bone slices (Fig 2m and n in II). The

α2,3 and α2,6 sialic acids containing protein, such as OPN, present at the resorption

pits decreased osteoclastogenesis on already resorbed bone slices.

5.3 Immunohistology of OPN and TRAcP in the areas of increased

bone metabolism in OA (II)

To relate the in vitro data of how the osteoclasts produce OPN during bone

resorption to in vivo conditions, immunohistochemistry was conducted to show the

presence of OPN and TRAcP in the areas of increased bone metabolism and

damage in osteoarthritic human cadaver femoral head samples.

Altogether biopsies were acquired from 32 femoral head samples. The biopsies

were taken from the damaged cartilage and sclerotic bone underneath it, trabecular

bone and cortical bone of the femoral neck. The samples were stained using an

EnVision Detection Systems Peroxidase/DAB, Rabbit/Mouse, HRP. Rabbit/Mouse

(DAB+) kit according to the manufacturer’s instructions and the samples were

visually quantified under a light microscope.

OPN was present in the extracellular matrix of all bone areas. However, the

staining was most intense in the osteoarthritic sclerotic bone areas, where bone

metabolism is increased, as expected, due to increased osteoclast activity (Table 2

in II).

Two different TRAcP antibodies were used. One specific for TRAcP 5A and

the other binding both 5B and 5A forms. TRAcP was also found in the extracellular

bone matrix. Both TRAcP antibodies stained the cortical bone slightly more than

sclerotic bone or trabecular bone, as shown in figure 9 (Table 2 in II). No

differences between the antibodies were seen in bone.

Page 65: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

63

Fig. 9. TRAcP 5A + 5B localization in the cortical bone (A) in the femoral neck and in the

trabecular bone (B) in the OA femoral head.

Page 66: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

64

Interestingly, as the samples also contained sections of damaged cartilage, staining

of the tight junction line between the non-calcified and the calcified cartilage was

TRAcP positive (Fig 4c and d in II). The staining was equally strong with both

TRAcP antibodies. When using the OPN antibody the line itself did not stain, but

intense staining was found in the calcified cartilage (Fig 4b in II). In non-calcified

cartilage the OPN stain would fade out further away from the tight junction. As a

control the staining protocol was done without the primary antibodies. No

unspecific staining was seen, as shown in figure 10.

Fig. 10. OA femoral head section section staining without primary antibody. No

unspecific staining was seen in cartilage, bone or bone marrow.

5.4 Analyses of the effect of RA serum and synovial fluid on

osteoclastogenesis and bone resorption (III)

To evaluate the combined effect of all inflammatory factors present in RA patients’

serum and synovial fluid samples, human peripheral blood derived osteoclasts were

cultured with them to study their effect on osteoclastogenesis and bone resorption.

Page 67: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

65

Healthy volunteers’ serum and OA patients’ synovial fluid were used as controls.

The number of osteoclasts on bone slices was counted after the culture period and

their morphology analyzed under a light microscope, and the resorbed volume of

bone slices was analyzed with a laser microscope.

In study I, OPN and its phosphorylation were analyzed with TRAcP 5A/5B

form concentrations. In study III, a multiplex analysis was conducted to quantify

other inflammatory factors in RA and OA that affect osteoclasts.

The number of samples used for each study are shown in table 3. The synovial

fluids were pooled due to the low volume of samples available and known high

variation of quality and biological diversity, to obtain standardized RA and OA

synovial fluids that represents the diseases in general and not the situation only

within a single joint.

Table 4. Patient samples analyzed in the study III.

Study Sample n

Osteoclast culture (Fig 1, 2 and Table 1 in III) seropositive RA patient synovial fluid 3 pooled

OA synovial fluid 3 pooled

RA serum 3

healthy serum 3

Multicytokine assay (Fig 3 and 4 and Supplement 1 in III) seropositive RA patient synovial fluid

OA patient synovial fluid

10

10

RA serum 6

Healthy serum 9

5.4.1 Effect of novel untreated RA patient serum on

osteoclastogenesis and bone resorption

Untreated RA patient serum increased the number of osteoclasts and volume of

resorbed bone significantly when compared to healthy control serum (Fig 1A in III).

The number of osteoclasts and the volume of resorption increased in the same ratio,

(~1.5 times) indicating that the increase in resorption was a direct result of

increased osteoclastogenesis (Fig 1C in III). No differences were seen in osteoclast

morphology in cultures with serums.

Page 68: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

66

5.4.2 Effect of RA and OA patients’ synovial fluid on

osteoclastogenesis and bone resorption

RA synovial fluid increased the osteoclastogenesis (~6 times) and the volume of

resorbed bone (~8 times) significantly when compared to healthy serum (Fig 1B

and D in III). Interestingly, OA synovial fluid increased the number of osteoclasts

and the resorption significantly more than RA synovial fluid (Fig 1B and D in III).

OA roughly doubled the number of osteoclasts and resorption, when compared to

RA. As in the serum assay, the increases in the bone resorption were a direct result

of the increased number of osteoclasts.

Synovial fluid changed the osteoclasts’ morphology (Fig 2 and Table 1 in III).

Both synovial fluids increased the osteoclasts’ size and the number of nuclei

significantly when compared to the healthy serum. However, the cells cultured with

OA synovial fluid were even larger, had more nuclei and were irregularly shaped

when compared to cells cultured with RA synovial fluid. On average the cells

cultured with healthy serum had two or three nuclei, the ones cultured with RA

synovial fluid had four to seven nuclei and the ones cultured in the presence of OA

synovial fluid had more than eight nuclei. Interestingly, the change in phenotype

did not affect the osteoclasts’ resorption capacity.

5.4.3 Cytokine analyses of the patient samples

To distinguish the factors behind the changes seen in the osteoclastogenesis and the

bone resorption 27 different cytokine and related protein concentrations (IL-1-β,

IL-1 receptor antagonist (IL-1ra), IL-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10,

IL-12, IL-13, IL-15, IL-17, eotaxin, FGF, granulocyte colony stimulating factor (G-

CSF), granulocyte-macrophage colony stimulating factor (Gm-CSF), IFN-γ, IP-10,

MCP-1, MIP-1-α, MIP-1-β, platelet-derived growth factor (PDGF-BB), RANTES,

TNF-α, and VEGF) were analyzed using a multicytokine assay kit from the

synovial fluids and serums. RANKL and OPG concentrations were also analyzed

from the synovial fluid samples.

In RA serum multiple cytokines associated with RA were found increased,

many of which are associated with increased osteoclastogenesis, thus explaining

the cell culture result. Significantly increased concentrations of IL-6 (3.5 fold

increase), IFN-gamma (1.5 fold increase), IP-10 (4.5 fold increase) and IL-9 (4.5

fold increase) were found in RA serum, and other cytokines that were considerably

increased in RA serum but did not reach statistical significance due to the wide

Page 69: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

67

distribution of results were IL-1ra (5 fold increase), TNF-α (2 fold increase),

RANTES (3 fold increase), IL-1b (5.5 fold increase), IL 13 (3 fold increase), MCP1

(4.5 fold increase) and IL-7 (5.5 fold increase) (Fig 3 and 4 in III).

In both OA and RA synovial fluids all tested cytokine concentrations were

generally increased when compared to either tested serum. When comparing the

synovial fluids, an increase of almost all cytokines in RA was found. As expected,

the most increased cytokines were associated with RA and increased

osteoclastogenesis, as in serum samples. Statistically significantly increased

cytokines between RA and OA synovial fluids were IL-1ra (85 fold increase), IL-

1b (10 fold increase), MCP-1 (30 fold increase), IL-8 (70 fold increase), IL-10 (15

fold increase), IL-15 (10 fold increase) and IL-17 (10 fold increase), and other

cytokines that were considerably increased but did not reach statistical significance

due to the wide variation in the measurements were MIP1b (30 fold increase), IL-

6 (10 fold increase), IFN-gamma (10 fold increase), GMCSf (10 fold increase),

TNF-α ( 30 fold increase), RANTES (5 fold increase) and IP-10 (40 fold increase)

(Fig 3 and 4 in III).

It is noteworthy, that in the serum samples the mean changes in the cytokine

concentrations were approximately one to six-fold, while in the synovial fluids the

changes in the cytokine concentrations were considerably larger, from five to

eighty-fold.

No differences were found in OPG concentrations in the synovial fluids of RA

and OA patients. Unfortunately, in all samples, all but one RA RANKL measure

fell under a detection level of 7.04pg/ml. However, this indicates that the 20ng/ml

concentration used in cell cultures is supraphysiological. Thus, the natural RANKL

in the samples does not cause confounding effect and the differences seen in the

osteoclast differentiation and the bone resorption are a result of other cytokines

present in the samples.

Page 70: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

68

Page 71: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

69

6 Discussion

During this millennium the interaction between the bone and the immune system

has proven to be crucial in the pathogenesis of RA and OA. New discoveries in the

field of osteoimmunology have led to the rapid development of new biological RA

drugs, which have proven very effective. However, these diseases are still often

debilitating for the patients and cause an enormous socioeconomical burden. New

treatment targets and treatments are sought rigorously to improve the patients’

quality of life.

In this thesis osteoclast function and factors that affect it in RA and OA were

studied. Especially OPN and its phosphorylation, which is controlled by TRAcP,

was studied in detail. Also, to understand the function of the osteoclasts in disease

one must better understand their function in the normal environment, hence basic

research on bone resorption by osteoclasts in a normal controlled environment was

conducted.

The phosphorylation of OPN was found to be increased in RA in study I, which

may lead to increased inflammatory cell and osteoclast activation along with the

other inflammatory factors, as shown in study III. In study II, the deposition of

OPN into the resorption pits on human bone by human osteoclasts was shown. In

the resorption pit OPN may affect mineralization and act as a signal for other cells

in the normal situation, but also in disease.

The results are discussed in detail in the next chapters. The clinical significance

of these new discoveries is to be discovered in future studies.

6.1 Decreased TRAcP 5B/5A ratio leads to increased

phosphorylation of OPN in RA synovial fluid.

In study I, the TRAcP 5B/5A isoenzyme concentration ratio’s relation to the OPN’s

phosphorylation was assessed in OA and RA synovial fluid. It was evident that the

decreased TRAcP 5B/5A ratio correlated with the increased phosphorylation of

OPN in RA. Earlier increased levels of OPN have been associated with a more

severe inflammatory disease phenotype (Gao et al., 2010; Iwadate et al., 2013; Shio

et al., 2010). However, the phosphorylation of OPN in inflammatory diseases has

not been studied before, even though it is the key regulator of OPN’s functions,

such as its role as a proinflammatory cytokine or inhibitor of mineralization

(Addison, Masica, Gray, & McKee, 2010; Ek-Rylander & Andersson, 2010;

Nyström et al., 2007; Sainger et al., 2012; L. Wang et al., 2008; Weber et al., 2002).

Page 72: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

70

OA patient synovial fluid was used as a control because acquiring synovial

fluid from healthy individuals is thought unethical and is very difficult due to the

low volume of fluid in a healthy joint. However, in a single study OPN’s

phosphorylation has been compared between synovial fluids of OA patients and

healthy controls, and shown that OPN is more phosphorylated in OA synovial fluid

(Xu et al., 2013). In respect to this study, according to our results it can be said that

OPN’s phosphorylation is significantly higher in RA synovial fluid than in healthy

synovial fluid.

TRAcP is the main regulator of extracellular OPN’s phosphorylation level and

thus the main regulator of its function, as it is believed, that OPN is secreted in its

phosphorylated form after being phosphorylated by intracellular kinases, such as

Fam20C (Andersson et al., 2003; Christensen et al., 2005). However, Fam20C

kinases have also been shown to be secreted extracellularly (Tagliabracci et al.,

2012). The combined effect of TRAcP and extracellular kinases has not been

studied. Total absence of Fam20C kinase leads to a rare disease called Raine

syndrome, which is characterized by congenital sclerosing osteomalacia and

cerebral calcification, which are partly thought to result from the loss of

biomineralizationinhibitory factors, such as phosphorylation of OPN (Whyte et al.,

2017).

TRAcP production has been shown to be elevated in both RA and OA (Janckila

et al., 2008; Seol et al., 2009). We did not find a difference in the overall TRAcP

concentrations between the diseases, but the TRAcP 5B/5A ratio was lower in RA

synovial fluid when compared to OA, and the decreased ratio correlated

significantly with the increased phosphorylation of OPN. We hypothesize that in

RA the production of phosphorylated OPN is upregulated during inflammation

(Lund, Giachelli, & Scatena, 2009), and insufficient TRAcP 5B levels in the

synovial fluid are not enough to dephosphorylate it to the normal level. The

increased phosphorylation of OPN in the synovial fluid leads to increased

inflammatory cell and osteoclasts activation, which leads to increased synovitis and

bone destruction, thus playing a part in the vicious circle of chronic inflammation

(Fig 4 in I).

It is worth mentioning that the synovial fluid and tissue samples were collected

during a knee prosthesis operation from patients suffering joint destruction, so the

duration of the chronic inflammation is rather long in both disease groups and this

situation may be different from the early stages of the diseases. The late stage

cytokine profile of the OA synovial fluid may resemble more RA than in the early

Page 73: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

71

stages of OA. However, the heterogeneity of the samples was reduced as the

patients must have met strict criteria for the knee prosthesis operation.

Earlier, TRAcP positive mono- and multinucleated cells have been found in the

OA and RA synovial tissue (Yamaguchi et al., 2014; Tsuboi et al., 2003). We did

not detect clearly multinucleated cells in our histological samples of OA or RA

synovial tissue, but we found that the synovial lining layer cells were strongly

TRAcP positive. An antibody detecting both TRAcP 5A and 5B forms, and another

antibody detecting only the 5A form was used. The staining in the lining layer was

identical with both antibodies and we suggest that most of the TRAcP in the lining

layer cells is in the less active 5A form. A slightly stronger staining with the TRAcP

antibody binding both forms was observed in OA than in RA samples, which may

indicate that there is more TRAcP 5B in the area, in accordance with the measured

TRAcP 5B/5A ratios in the synovial fluid, but further studies of TRAcP expression

in the synovial tissue are needed.

TRAcP 5A is proteolytically processed into the 5B form by cathepsin K and

other proteinases (Fagerlund et al., 2006; Lång & Andersson, 2005; Ljusberg et al.,

2005). Interestingly, in earlier studies the levels of these proteinases have been

shown to be higher in RA than in OA synovial tissue or fluid (Hou et al., 2002;

Pozgan et al., 2010). In relation to the data, it seems possible that some mechanism

may prevent TRAcP 5A from being processed in RA into the 5B form or the

conversion mechanism does not function properly. In future studies it would be

interesting to further look into what is the cell in the synovial tissue that produces

TRAcP. It was speculated that most of the TRAcP positive cells in the synovial

lining are synovial macrophages, but the general staining of the cells suggests that

synovial fibroblasts could also be TRAcP positive. This raises a question on the

role of TRAcP in fibroblast-like synoviocytes and its origin. It has been suggested

that the synovial cells can differentiate into bone resorbing cells under the right

circumstances (Suzuki et al., 2001), but as the standard synovial cells are mostly

TRAcP 5A positive, not 5B positive as are the osteoclasts, synovial cells are

unlikely to resorb bone or cartilage themselves.

6.2 OPN is one of the key proteins secreted into the resorption

lacunae during bone resorption

In study II, the secretion of OPN into the resorption lacunae by human osteoclasts

during bone resorption was verified and different glycan epitopes at the bottom of

the resorption pit were identified. Additionally, the resorption and deposition of OPN

Page 74: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

72

on pre-deposited protein free carbonated hydroxyapatite was also analyzed. The

presence of OPN and its phosphatase TRAcP was also evaluated histologically

from cartilage and subchondral bone biopsies of patients suffering from OA to

show their presence in the areas of increased mineral metabolism.

Various diseases and situations affect extracellular OPN levels and post

translational modifications, such as phosphorylation, as shown in study I. The

effects of exogenous extracellular OPN on osteoclasts are better understood than

the function of endogenous osteoclast OPN. Previous studies have investigated the

secretion of OPN into the resorption lacunae during bone resorption but have used

either animal cells or bone (Dodds et al., 1995; Maeda et al., 1994; Shimazu, Nanci,

& Aoba, 2002). In study II, the deposition of OPN by osteoclasts was investigated for

the first time using human cells on human bone and analyzed with modern FE-SEM

methods. The main discovery of this study was that OPN is specficly secreted by both

human bone marrow and peripheral blood derived osteoclasts into the resorption

lacunae on human bone and on protein-free carbonated hydroxyapatite. The proteomics

analyses of the resorbed bone slices showed that OPN was the most increased protein

in the resorbed bone. The glycan epitope analyses from the bottom of resorption lacunae

showed an increase of α2,3 and α2,6 -linked sialic acids, which were mostly in O-

glycans. This supports the discovery that OPN is the most abundant protein secreted

into the resorption pit, as the other enriched proteins do not contain both of the

sialic acid residues. Also, as the mineral content and crystallinity of bone in OPN

knockout mice is increased, this supports the hypothesis that OPN has a key role in

the regulation of bone metabolism, by likely acting as an important mediator of

activity for both the osteoclasts and the osteoblasts (Boskey et al., 2002).

The proteins secreted into the resorption pit function as signals for other bone

cells; osteoclasts, osteoblasts and osteocytes (H. Li et al., 2016; Lotinun et al.,

2013). Osteoclasts are thought to attach themselves to bone during resorption

through the αvβ3-integrin receptor, which is shown to be able to bind OPN’s RGD-

sequence (Ek-Rylander & Andersson, 2010; Ross et al., 1993; Sodek et al., 2000).

We support this hypothesis, as we found that OPN is secreted into the resorption

pit also on carbonated hydroxyapatite, and it has been previously shown that OPN

can bind to hydroxyapatite (Goldberg, Warner, Li, & Hunter, 2001). Thus, our

suggestion is that OPN acts as a connecting molecule between the resorbed

hydroxyapatite and the cell surface receptor.

TRAcP is the primary osteoclast marker, and it has also been shown to be

secreted into the resorption lacunae during bone resorption (Andersson et al.,

2003). Since OPN’s ability to inhibit biomineralization is dependent on its

Page 75: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

73

phosphorylation (Gericke et al., 2005; Hoac et al., 2017; Wang et al., 2008), it would

seem paradoxical that OPN is dephosphorylated by TRAcP in the resorption pit during

the resorption. And, it has been shown that osteoclasts’ migration towards OPN is

dependent on its phosphorylation (Ek-Rylander & Andersson, 2010). We suggest that

OPN may be dephosphorylated during or at the late stage of resorption in order to mark

the resorbed area not to be resorbed again by other osteoclasts.

Extracellular OPN’s effects on osteoblasts are not fully understood (Icer &

Gezmen-Karadag, 2018). However, various other signals secreted by osteoclasts

that affect osteoblasts, the so-called clastokines, are known: they either increase or

decrease osteoblast activity (Cappariello et al., 2014). A phosphorylated form of

OPN is suggested to decrease osteoblast activity in bone, and, thus, have also cell

mediated inhibitory effects on the mineralization (Holm et al., 2014; Kusuyama et

al., 2017). However, only a single study so far has studied the effect of

phosphorylated and de-phosphorylated OPN on osteoblasts. On SaOS-2 cells, an

osteoblast-like cell line, phosphorylated OPN decreased their mineralization

activity but de-phosphorylated OPN did not (Halling Linder et al., 2017). Our

research group’s hypothesis was, that OPN is dephosphorylated during bone

resorption to enable new mineralization by osteoblasts.

It is known that the resorbed area on bovine bone can be recognized using

WGA-lectin (Selander, Lehenkari, & Vaananen, 1994), which binds to β1,4-linked

N-acetylglucosamine (Allen, Neuberger, & Sharon, 1973). In this study, WGA and

multiple other lectins were used to study the glycan epitope in the the resorption pits

on human bone. Strong binding of lectins indicating an abundance of α2,3- and α2,6-

linked sialic acids was found in the resorption lacunae, with most of the sialic residues

in O-glycans. This is consistent with the hypothesis that the major protein found at the

bottom of the resorption pit is OPN. When the sialic residues were enzymatically

removed from the resorbed bone slices and osteoclasts re-cultured on them, the number

of osteoclasts doubled when compared to untreated pre-resorbed bone slices. This may

mark that osteoclastogenesis is increased because more α2,3/6-linked sialic acid or a

protein containing it, such as OPN, is required on the bone. Or the protein’s inhibitory

effect on osteoclast differentiation is lost after de-sialylation.

It has been suggested that exogenous OPN may actually inhibit osteoclastogenesis

in certain conditions, but during osteoclastogenesis from monocytes, OPN expression

is increased, and it still acts as an important mediator of migration and activity (Ge et

al., 2017). As discussed earlier, the hypothesis is that osteoclasts may deposit OPN, or

another protein containing the above sialic residues, on bone to act as marker in

resorbed areas for other osteoclasts not to resorb the same areas again. This effect may

Page 76: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

74

be dependent on the level OPN’s phosphorylation, which would be interesting to test

in future studies.

6.3 OPN is found in the calcified cartilage

In study II, immunohistochemistry of OPN, and TRAcP was conducted to

investigate their presence in areas of increased bone metabolism in OA. Areas from

subchondral, trabecular and cortical bone were analyzed. OPN staining was slightly

stronger in the subchondral sclerotic bone, than in the other areas. In general, the

bone metabolism is thought to be increased in the sclerotic bone. However, the

staining for OPN was the most intense in the calcified cartilage, a transition area

between the non-calcified cartilage and the subchondral bone. The line between

normal cartilage and calcified cartilage is called the tidemark.

Both TRAcP antibodies stained the cortical bone slightly more than the

sclerotic or trabecular bone. No increase of TRAcP in the areas of increased bone

metabolism in the samples was seen. No differences in bone staining between the

antibody specific for 5A or the other binding both 5A and 5B forms were seen.

Interestingly, both TRAcP antibodies used showed a strong staining of the

cartilage tidemark, while no TRAcP was detected in either calcified cartilage or

normal cartilage. The presence of TRAcP in the tidemark may indicate a change in

the phosphorylation of OPN between normal and calcified cartilage. Our

hypothesis is, that TRAcP dephosphorylates OPN at the tidemark, which causes its

inhibitory effect on biomineralization to be lost (Gericke et al., 2005; Hoac et al.,

2017; L. Wang et al., 2008), which leads to increased calcification of the cartilage

at the bone side of the tidemark, the calcified cartilage. Unfortunately, the

phosphorylation of OPN cannot be easily quantified histologically, as there is no

specific antibody available to detect phosphorylated or de-phosphorylated OPN.

6.4 RA synovial fluid and serum increase osteoclastogenesis and

bone resorption in vitro

In study III, the effect of inflammatory factors present in RA synovial fluid and

serum on osteoclast differentiation and bone resorption was investigated. OA

synovial fluid and healthy serum were used as controls. As in study I, OA patients’

synovial fluid was used as a control due to ethical issues with acquiring healthy

controls’ synovial fluid. Supraphysiological RANKL concentrations were used in

all cell cultures to elucidate the effect of other factors affecting the

Page 77: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

75

osteoclastogenesis. It was evident that RA synovial fluid and serum both increased

the osteoclast differentiation, when compared to healthy controls. Surprisingly OA

synovial fluid increased the osteoclastogenesis even more than RA synovial fluid.

Inflammatory cytokines from the serums and synovial fluids were analyzed

with a 27-cytokine multiplex assay. A general increase of the inflammatory

cytokines was found between RA and healthy control serums, and RA and OA

synovial fluids. Surprisingly, the higher concentrations of inflammatory cytokines

in RA synovial fluid did not lead to increased osteoclastogenesis and activity when

compared to OA. However, morphological differences between the osteoclasts

cultured with different synovial fluids were found, as the cells cultured with OA

synovial fluid were significantly larger and contained more nuclei. No differences

in the osteoclast morphology were seen in cultures with serum samples.

Previous studies of osteoclastogenesis in the areas of inflammatory bone

erosions in RA have suggested that the pro-inflammatory cytokines present in RA

synovial tissue, along with increased RANKL stimulus, cause so-called

inflammatory osteoclastogenesis in the inflamed RA synovial tissue adjacent to

bone (Schett & Gravallese, 2012; Suzuki et al., 2001; Tsuboi et al., 2003). However,

the osteoclasts generated by the inflammatory osteoclastogenesis have been

described to be smaller and contain fewer nuclei, and thus present a more

macrophage like phenotype, than the osteoclasts in normal bone (Suzuki et al., 2001;

Tsuboi et al., 2003). We believe that in our experiment with the synovial fluids the

strong pro-inflammatory stimulus present in RA synovial fluid causes the

osteoclasts to stay in a more macrophage like phenotype than in the cultures with

OA synovial fluid, which seemed to create a very good environment for the

osteoclastogenesis. Interestingly, when comparing the volume of bone resorbed by

a single osteoclast the change in morphology did not affect it. The change in

morphology may, however, affect the osteoclasts’ interaction with the other bone

cells, and this may partly explain why the changes in the bone metabolism between

RA and OA happen in the different areas of the joint.

RANKL is the main factor of osteoclastogenesis, along with M-CSF, which

first activates the production of the RANK cell surface receptor, as explained in

chapter 2.1.3 (Boyce, 2013). Our data are in accordance with RANKL signaling

being the most crucial for osteoclastogenesis, as deprivation of it in negative control

samples inhibited the osteoclast differentiation. To evaluate the effect of other

factors affecting osteoclastogenesis, the effect of RANKL already present in the

samples was nullified by using supraphysiological RANKL concentrations, and

synovial fluid RANKL and OPG levels were measured to avoid confounding by

Page 78: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

76

the natural RANKL and OPG in them. Earlier studies have assessed various

cytokines that promote osteoclastogenesis via RANKL or other pathways. These

cytokines include TNF-α, IL-1, IL-6, IL-7, IL-8, IL-11, IL-15, IL-17, IL-23, and

IL-34 (Amarasekara et al., 2018). Of the cytokines associated with increased

osteoclastogenesis, we found increased levels of TNF-α, IL-1, IL-6, IL-7, IL-8, IL-

15, IL-17 in our RA patient samples, IL-11 and IL-23 were not tested. Also, various

other disease and inflammation related molecules and proteins, such as, ACPAs,

CRP and VEGF in RA, are known to increase osteoclast differentiation

independently of RANKL (Harre et al., 2015; H. Kim et al., 2015; K. Kim et al.,

2015). An increase of VEGF was seen in the RA synovial fluid samples. CRP was

not tested. All the RA patients selected for the study were ACPA positive.

However, all cytokines are not pro-inflammatory and increase

osteoclastogenesis: various anti-inflammatory cytokines are known to be increased

in both RA and OA. Cytokines that are associated with decreased osteoclast

differentiation include IFN-α, IFN-β, IFN-γ, IL-3, IL-4, IL-10, IL-12, IL-27 and

IL-33 (Amarasekara et al., 2018). Of the above we found IFN-γ, IL-4 and IL-10 to

be increased in the tested RA samples. Due to limitations in the number of cytokines

tested in the multiplex assay, the other anti-osteoclastic cytokines were not tested.

However, previous studies have found increased concentrations of IFN-α, IL-3, IL-

27 and IL-33 in RA patient samples (Ali Khan, 2018; Lai et al., 2016; Pavlovic et

al., 2016; Sellam et al., 2016).

It is evident that the combined cytokine effect in our cell culture experiment

favours osteoclastogenesis, as could be seen from the osteoclast cultures with RA

serum or either synovial fluid compared to healthy control serum. The

inflammatory stimulus present in both synovial fluid and serum increased the

osteoclastogenesis significantly, but apparently the effect is limited, as OA synovial

fluid, which contained less pro-inflammatory factors increased the osteoclast

differentiation significantly more than RA. Since the study samples are from real

patients, we believe that this depicts rather well the complex situation in the

diseased joint where both pro- and anti-inflammatory factors work at the same time.

As discussed earlier, the unexpected result seen between the synovial fluids is likely

caused by the inflammatory osteoclastogenesis, which drives the osteoclasts

towards a more macrophage-like phenotype (Suzuki et al., 2001; Tsuboi et al.,

2003). It is notable that the RA serum samples were collected from untreated RA

patients at the time of diagnosis and the synovial fluid samples during knee

prothesis operation from RA patients suffering from secondary arthrosis of the knee.

This limitation is due to the availability of patient material. However, the data

Page 79: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

77

shows, that a significant inflammatotry factors exist in the synovial fluid of RA

patients with secondary arthrosis of the knee, which etiology differs from that of

primary OA.

RA and OA are both diseases which are associated with both local and systemic

changes in the bone metabolism specific for the disease (Greisen et al., 2015;

Prieto-Potin, Largo, Roman-Blas, Herrero-Beaumont, & Walsh, 2015; Suzuki et al.,

2001). In RA, the inflamed synovial tissue is the main source of inflammatory

cytokines, including RANKL (Tsuboi et al., 2003), and the bone erosions start from

the perichondral areas where the synovial tissue is in contact with the bone, which

is not covered by the articular cartilage. In OA, the mechanical wear of the cartilage

and the subchondral bone along with the synovial inflammation cause changes in

local bone metabolism, which results in multiple pathological changes, such as

sclerosis, cysts and osteophytes, all of which are also associated with increased

osteoclastic activity (Barr et al., 2015; Favero et al., 2015).

The bone erosions in RA are caused by osteoclasts (Cappariello et al., 2014).

According to our data we suggest that the monocytes or macrophages in the

inflamed synovial tissue differentiate into osteoclasts due to inflammatory

osteoclastogenesis and cause the inflammatory bone erosions in RA. According to

the data in study I, the synovial cells are unlikely to resorb bone themselves as they

present a more macrophage than osteoclastic phenotype, as they are TRAcP 5A,

not 5B positive. However, it has been suggested that under the right conditions it

may be possible for them to undergo changes that would allow them to fuse into

bone resorbing cells (Suzuki et al., 2001). RA is also associated with an increased

risk of osteoporosis, which is thought to be a result of both medication and

circulating inflammatory cytokines filtered into the serum from the inflamed

synovial tissue (Clayton & Hochberg, 2013). OA is not associated with a risk of

increased osteoporosis (Clayton & Hochberg, 2013). Our osteoclast culture

experiment with RA serum shows how the circulating inflammatory cytokines in

RA increase the osteoclastic activity, and hence the risk of general osteoporosis.

Our cytokine data is in accordance with the previous literature of cytokines in

RA and OA, where the severity of both diseases has been shown to correlate with

the increased cytokine levels (Barra et al., 2014; Hampel, Sesselmann, Iserovich,

Sel, Paulsen, & Sack, 2013; Vangsness, Burke, Narvy, MacPhee, & Fedenko, 2011).

In the synovial fluid and serum, the level of the major pro-inflammatory cytokines

associated with RA, such as Il-1, Il-6, IL-8, VEGF etc., were increased as expected.

In general, the measured cytokines were ten times more concentrated in the

synovial fluids than in the serums, which was also seen in the fold changes. The

Page 80: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

78

changes in the cytokines, which reached statistically significant differences,

between serum and synovial fluid experiments are explained by the low number of

study subjects and measurements of certain serum cytokines falling under the

detection level in the multicytokine assay.

In conclusion, OA and RA fluids affect the osteoclastogenesis and the bone

resorption by osteoclasts in vitro. Both RA synovial fluid and serum increased the

osteoclastogenesis and the bone resorption. Surprisingly, the osteoclastogenesis

and the bone resorption increased the most in the presence of OA patient synovial

fluid. The bone changes happening in the diseases can better understood with these

data. However, further studies of inflammatory osteoclastogenesis are required to

obtain better understanding for optimal therapeutic interventions for the

inflammatory diseases affecting joints and bone. This in vitro disease cell model

could be used to evaluate the effect of new potential drugs on osteoclast function.

6.5 Future aspects

The observation that the TRAcP 5B/5A ratio differs and affects OPN’s

phosphorylation in RA raises questions about the mechanism behind this. TRAcP

positive cells were abundant in the synovial tissue. In future studies it would be

interesting to study the expression of TRAcP in the synovial cells in detail to

distinguish the cell type that produces it. The mechanism behind the change in the

TRAcP 5B/5A ratio between RA and OA synovial fluids would also be interesting

to study further, as discussed earlier in chapter 6.1. It seemed possible, that the

proteolytic mechanism behind TRAcP 5A being processed to 5B does not work in

RA, as the total TRAcP levels did not differ between RA and OA.

As the other factor that affects OPN’s phosphorylation, the in vivo relevance

of extracellular Fam20C kinase on OPN’s phosphorylation should also be studied

from real patient samples, along with its relation to TRAcP. Which affects OPN’s

phosphorylation more?

The effect of extracellular OPN and its phosphorylation on osteoblasts should

be studied more, to gain further understanding of the osteoclast-osteoblast

connection. Better understanding of the basic functions of the bone cells may reveal

new therapeutical prospectives in diseases related to changes in bone metabolism,

such as osteoporosis.

To possibly reveal new insights in the pathogenesis of OA, it would be

interesting to measure OPN’s phosphorylation in the calcified cartilage of healthy

Page 81: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

79

and OA patients. The change in OPN’s phosphorylation may be an important factor

in the changes seen in the OA cartilage and the subchondral bone.

Inflammatory osteoclastogenesis is an interesting phenomenon and according

to our results the osteoclasts may differ from the normal osteoclasts. The

inflammatory osteoclastogenesis is not yet well understood. In future studies, the

inflammatory osteoclasts should be studied to reveal further differences, which

could possibly be influenced with targeted therapies. The therapies against

inflammatory osteoclastogenesis would allow us to affect only the pathological

osteoclasts, without affecting the normal bone metabolism.

Page 82: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

80

Page 83: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

81

7 Conclusions

The main goal of this study was to investigate the osteoclast function and the factors

that affect it in normal or pathological conditions of RA and OA. Based on the

study’s results, the following major conclusions were made.

1. The phosphorylation of OPN is increased in RA synovial fluid, which is the

result of the decreased TRAcP 5B/5A ratio.

2. Human osteoclasts secrete OPN into the resorption pit during the resorption of

human bone or a protein free resorbable material. OPN is the most abundant

protein secreted during the bone resorption into the resorption area.

3. RA serum and synovial fluid increased the osteoclastogenesis and the bone

resorption when compared to the healthy control serum. However, OA synovial

fluid increased the osteoclastogenesis and activity even more. RA synovial

fluid contains high concentrations of inflammatory cytokines. These cause the

inflammatory osteoclastogenesis, which generates osteoclasts of a more

macrophage like phenotype.

These interesting new discoveries will reveal their clinical significance in future

studies.

Page 84: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

82

Page 85: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

83

List of references

Addison, W. N., Masica, D. L., Gray, J. J., & McKee, M. D. (2010). Phosphorylation-dependent inhibition of mineralization by osteopontin ASARM peptides is regulated by PHEX cleavage. Journal of Bone & Mineral Research, 25(4), 695-705. doi:10.1359/jbmr.090832

Aho, K., Kaipiainen-Seppänen, O., Heliövaara, M., & Klaukka, T. (1998). Epidemiology of rheumatoid arthritis in Finland. Seminars in Arthritis and Rheumatism, 27(5), 325-334. doi:10.1016/S0049-0172(98)80053-X

Aletaha, D., Neogi, T., Silman, A. J., Funovits, J., Felson, D. T., Bingham III, C. O., . . . Hawker, G. (2010). 2010 rheumatoid arthritis classification criteria: An american college of Rheumatology/European league against rheumatism collaborative initiative. Arthritis & Rheumatism, 62(9), 2569-2581. doi:10.1002/art.27584

Ali Khan, W. (2018). Recombinant interferon alpha 2b in rheumatoid arthritis: Good antigen for rheumatoid arthritis antibodies. Central European Journal of Immunology, 43(1), 58-68. doi:10.5114/ceji.2018.74874

Allen, A. K., Neuberger, A., & Sharon, N. (1973). The purification, composition and specificity of wheat-germ agglutinin. Biochemical Journal, 131(1), 155-162. doi:10.1042/bj1310155

Alliston, T., Hernandez, C. J., Findlay, D. M., Felson, D. T., & Kennedy, O. D. (2018). Bone marrow lesions in osteoarthritis: What lies beneath. Journal of Orthopaedic Research, 36(7), 1818-1825. doi:10.1002/jor.23844

Amarasekara, D. S., Yun, H., Kim, S., Lee, N., Kim, H., & Rho, J. (2018). Regulation of osteoclast differentiation by cytokine networks. Immune Network, 18(1), e8. doi:10.4110/in.2018.18.e8

An, G., Acharya, C., Feng, X., Wen, K., Zhong, M., Zhang, L., . . . Anderson, K. C. (2016). Osteoclasts promote immune suppressive microenvironment in multiple myeloma: Therapeutic implication. Blood, 128(12), 1590-1603. doi:10.1182/blood-2016-03-707547

Andersson, G., Ek-Rylander, B., Hollberg, K., Ljusberg-Sjölander, J., Lång, P., Norgård, M., . . . Zhang, S. (2003). TRACP as an osteopontin phosphatase. Journal of Bone and Mineral Research, 18(10), 1912-1915. doi:10.1359/jbmr.2003.18.10.1912

Arnett, T. R. (2010). Acidosis, hypoxia and bone. Archives of Biochemistry and Biophysics, 503(1), 103-109. doi:10.1016/j.abb.2010.07.021

Arokoski, J., Manninen, P., Kröger, H., Heliövaara, M., Nykyri, E., & Impivaara, O. (2007). Hip and knee pain and osteoarthritis. In L. Kaila-Kangas (Ed.), Musculoskeletal disorders and diseases in Finland. Results of the health 2000 survey (pp. 37-41)

Arron, J. R., & Choi, Y. (2000). Osteoimmunology: Bone versus immune system. Nature, 408(6812), 535-536.

Ashkar, S., Weber, G. F., Panoutsakopoulou, V., Sanchirico, M. E., Jansson, M., Zawaideh, S., . . . Cantor, H. (2000). Eta-1 (osteopontin): An early component of type-1 (cell-mediated) immunity. Science, 287(5454), 860-864. doi:10.1126/science.287.5454.860

Page 86: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

84

Bailey, A. J., Mansell, J. P., Sims, T. J., & Banse, X. (2004). Biochemical and mechanical properties of subchondral bone in osteoarthritis. Biorheology 41(3-4):349-358.

Barr, A. J., Campbell, T. M., Hopkinson, D., Kingsbury, S. R., Bowes, M. A., & Conaghan, P. G. (2015). A systematic review of the relationship between subchondral bone features, pain and structural pathology in peripheral joint osteoarthritis. Arthritis Research & Therapy, 17(1), 228. doi:10.1186/s13075-015-0735-x

Barra, L., Summers, K., Bell, D., & Cairns, E. (2014). Serum cytokine profile of unaffected first-degree relatives of patients with rheumatoid arthritis. The Journal of Rheumatology, 41(2), 280-285. doi:10.3899/jrheum.130539

Bazzazi, H., Aghaei, M., Memarian, A., Hossein Asgarian-Omran, Behnampour, N., & Yazdani, Y. (2018). Th1-Th17 ratio as a new insight in rheumatoid arthritis disease. Iranian Journal of Allergy, Asthma and Immunology, 17(1), 68-77.

Bennike, T., Ayturk, U., Haslauer, C. M., Froehlich, J. W., Proffen, B. L., Barnaby, O., . . . Steen, H. (2014). A normative study of the synovial fluid proteome from healthy porcine knee joints. Journal of Proteome Research, 13(10), 4377-4387. doi:10.1021/pr500587x

Berger, A. (2000). Th1 and Th2 responses: What are they? British Medical Journal, 321(7258), 424. doi:10.1136/bmj.321.7258.424

Bhattacharyya, S., Pal, S., & Chattopadhyay, N. (2018). Targeted inhibition of sclerostin for post-menopausal osteoporosis therapy: A critical assessment of the mechanism of action. European Journal of Pharmacology, 826, 39-47. doi:10.1016/j.ejphar.2018.02.028

Bhosale, A. M., & Richardson, J. B. (2008). Articular cartilage: Structure, injuries and review of management. British Medical Bulletin, 87(1), 77-95. doi:10.1093/bmb/ldn025

Bonewald, L. F., & Wacker, M. J. (2013). FGF23 production by osteocytes. Pediatric Nephrology (Berlin, Germany), 28(4), 563-568. doi:10.1007/s00467-012-2309-3

Boskey, A. L. (2013). Bone composition: Relationship to bone fragility and antiosteoporotic drug effects. BoneKEy Reports, 2, 447. doi:10.1038/bonekey.2013.181

Boskey, A. L., Spevak, L., Paschalis, E., Doty, S. B., & McKee, M. D. (2002). Osteopontin deficiency increases mineral content and mineral crystallinity in mouse bone. Calcified Tissue International, 71(2), 145-154. doi:10.1007/s00223-001-1121-z

Boyce, B. F. (2013). Advances in the regulation of osteoclasts and osteoclast functions. Journal of Dental Research, 92(10), 860-867. doi:10.1177/0022034513500306

Boyle, W. J., Simonet, W. S., & Lacey, D. L. (2003). Osteoclast differentiation and activation. Nature, 423, 337-342. doi:10.1038/nature01658

Briggs, A. M., Woolf, A. D., Dreinhöfer, K., Homb, N., Hoy, D. G., Kopansky-Giles, D., … March, L. (2018). Reducing the global burden of musculoskeletal conditions. Bulletin of the World Health Organization, 96(5), 366–368. doi:10.2471/BLT.17.204891

Brown, A. (2012). Osteopontin: A key link between immunity, inflammation and the central nervous system. Translational Neuroscience, 3(3), 288-293. doi:10.2478/s13380-012-0028-7

Page 87: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

85

Bucay, N., Sarosi, I., Dunstan, C. R., Morony, S., Tarpley, J., Capparelli, C., . . . Simonet, W. S. (1998). Osteoprotegerin-deficient mice develop early onset osteoporosis and arterial calcification. Genes & Development, 12(9), 1260-1268. doi:10.1101/gad.12.9.1260

Bullock, W. A., Pavalko, F. M., & Robling, A. G. (2019). Osteocytes and mechanical loading: The wnt connection. Orthodontics & Craniofacial Research, 22, 175-179. doi:10.1111/ocr.12282

Bune, A. J., Hayman, A., J Evans, M., & Cox, T. (2001). Mice lacking tartrate-resistant acid phosphatase (ACP5) have disordered macrophage inflammatory responses and reduced clearance of the pathogen, staphylococcus aureus. Immunology, 102(1), 103-113. doi:10.1046/j.1365-2567.2001.01145.x

Calabresi, E., Petrelli, F., Bonifacio, A. F., Puxeddu, I., & Alunno, A. (2018) One year in review 2018: Pathogenesis of rheumatoid arthritis. Clinical and Experimental Rheumatology 36(2), 175-184.

Cappariello, A., Maurizi, A., Veeriah, V., & Teti, A. (2014). The great beauty of the osteoclast. Archives of Biochemistry and Biophysics 558, 70-78. doi:10.1016/j.abb.2014.06.017

Capulli, M., Paone, R., & Rucci, N. (2014). Osteoblast and osteocyte: Games without frontiers. Archives of Biochemistry and Biophysics, 561, 3-12. doi:10.1016/j.abb.2014.05.003

Chao, T., Wu, Y., & Janckila, A. J. (2010). Tartrate-resistant acid phosphatase isoform 5b (TRACP 5b) as a serum maker for cancer with bone metastasis. Clinica Chimica Acta, 411(21), 1553-1564. doi:10.1016/j.cca.2010.06.027

Charles, J. F., & Aliprantis, A. O. (2014). Osteoclasts: More than ‘bone eaters’. Trends in Molecular Medicine 20(8), 449-459. doi:10.1016/j.molmed.2014.06.001

Chen, G., Zhang, X., Li, R., Fang, L., Niu, X., Zheng, Y., . . . Zhang, J. Z. (2010). Role of osteopontin in synovial Th17 differentiation in rheumatoid arthritis. Arthritis & Rheumatism, 62(10), 2900-2908. doi:10.1002/art.27603

Chen, H., Senda, T., & Kubo, K. (2015). The osteocyte plays multiple roles in bone remodeling and mineral homeostasis. Medical Molecular Morphology, 48(2), 61-68. doi:10.1007/s00795-015-0099-y

Chen, Y., Janckila, A., Chao, T., Yeh, R., Gao, H., Lee, S., . . . Dai, M. (2015). Association of tartrate-resistant acid phosphatase-expressed macrophages and metastatic breast cancer progression. Medicine, 94(48), e2165. doi:10.1097/MD.0000000000002165

Cheng, C., Gao, S., & Lei, G. (2014). Association of osteopontin with osteoarthritis. Rheumatology International, 34(12), 1627-1631. doi:10.1007/s00296-014-3036-9

Christensen, B., Nielsen, M. S., Haselmann, K. F., Petersen, T. E., & Sorensen, E. S. (2005). Post-translationally modified residues of native human osteopontin are located in clusters: Identification of 36 phosphorylation and five O-glycosylation sites and their biological implications. Biochemical Journal, 390(Pt 1), 285-292. doi:10.1042/BJ20050341

Page 88: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

86

Chung, C. P., Oeser, A., Raggi, P., Gebretsadik, T., Shintani, A. K., Sokka, T., . . . Stein, C. M. (2005). Increased coronary-artery atherosclerosis in rheumatoid arthritis: Relationship to disease duration and cardiovascular risk factors. Arthritis & Rheumatism, 52(10), 3045-3053. doi:10.1002/art.21288

Clarke, B. L., Drake, M. T., Oursler, M. J., & Khosla, S. (2017). Cathepsin K inhibitors for osteoporosis: Biology, potential clinical utility, and lessons learned. Endocrine Reviews, 38(4), 325-350. doi:10.1210/er.2015-1114

Clayton, E. S., & Hochberg, M. C. (2013). Osteoporosis and osteoarthritis, rheumatoid arthritis and spondylarthropathies. Current Osteoporosis Reports, 11(4), 257-262. doi:10.1007/s11914-013-0172-1

Costa-Rodrigues, J., Fernandes, A., & Fernandes, M. H. (2011). Spontaneous and induced osteoclastogenic behaviour of human peripheral blood mononuclear cells and their CD14+ and CD14− cell fractions. Cell Proliferation, 44(5), 410-419. doi:10.1111/j.1365-2184.2011.00768.x

Dadoun, S., Zeboulon-Ktorza, N., Combescure, C., Elhai, M., Rozenberg, S., Gossec, L., & Fautrel, B. (2013). Mortality in rheumatoid arthritis over the last fifty years: Systematic review and meta-analysis. Joint Bone Spine, 80(1), 29-33. doi:10.1016/j.jbspin.2012.02.005

Dar, H.Y, Azam, Z., Anupam, R., Mondal, R. K., & Srivastava, R. K. (2018). Osteoimmunology: The nexus between bone and immune system. Frontiers in Bioscience 23(3), 464-492. doi:10.2741/4600

Deane, K. D. (2014). Preclinical rheumatoid arthritis (autoantibodies): An updated review. Current Rheumatology Reports, 16(5), 419. doi:10.1007/s11926-014-0419-6

Delaisse, J. M. (2014). The reversal phase of the bone-remodeling cycle: Cellular prerequisites for coupling resorption and formation. BoneKEy Reports, 3, 561. doi:10.1038/bonekey.2014.56.eCollection 2014

Dimitroulas, T., Lambe, T., Klocke, R., Kitas, G. D., & Duarte, R. V. (2017). Biologic drugs as analgesics for the management of osteoarthritis. Seminars in Arthritis and Rheumatism, 46(6), 687-691. doi:10.1016/j.semarthrit.2016.12.001

Dodds, R. A., Connor, J. R., James, I. E., Lee Rykaczewski, E., Appelbaum, E., Dul, E., & Gowen, M. (1995). Human osteoclasts, not osteoblasts, deposit osteopontin onto resorption surfaces: An in vitro and ex vivo study of remodeling bone. Journal of Bone and Mineral Research, 10(11), 1666-1680. doi:10.1002/jbmr.5650101109

Dougall, W. C., Glaccum, M., Charrier, K., Rohrbach, K., Brasel, K., De Smedt, T., . . . Schuh, J. (1999). RANK is essential for osteoclast and lymph node development. Genes & Development, 13(18), 2412-2424. doi:10.1101/gad.13.18.2412

Ek-Rylander, B., & Andersson, G. (2010). Osteoclast migration on phosphorylated osteopontin is regulated by endogenous tartrate-resistant acid phosphatase. Experimental Cell Research, 316(3), 443-451. doi:10.1016/j.yexcr.2009.10.019

Esfandiari, E., Bailey, M., Stokes, C. R., Cox, T. M., Evans, M. J., & Hayman, A. R. (2006). TRACP influences Th1 pathways by affecting dendritic cell function. Journal of Bone and Mineral Research, 21(9), 1367-1376. doi:10.1359/jbmr.060611

Page 89: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

87

Fagerlund, K. M., Ylipahkala, H., Tiitinen, S. L., Janckila, A. J., Hamilton, S., Mäentausta, O., . . . Halleen, J. M. (2006). Effects of proteolysis and reduction on phosphatase and ROS-generating activity of human tartrate-resistant acid phosphatase. Archives of Biochemistry and Biophysics, 449(1-2), 1-7. doi:/10.1016/j.abb.2006.03.010

Favero, M., Giusti, A., Geusens, P., Goldring, S. R., Lems, W., Schett, G., & Bianchi, G. (2015). OsteoRheumatology: A new discipline?. RMD Open, 1(Suppl 1), e000083.

Florencio-Silva, R., Sasso, G. R.., Sasso-Cerri, E., Simões, M. J., & Cerri, P. S. (2015). Biology of bone tissue: Structure, function, and factors that influence bone cells. BioMed Research International, 2015, 421746.. doi:10.1155/2015/421746

Fortis, S., Khadaroo, R. G., Haitsma, J. J., & Zhang, H. (2015). Osteopontin is associated with inflammation and mortality in a mouse model of polymicrobial sepsis. Acta Anaesthesiologica Scandinavica, 59(2), 170-175. doi:10.1111/aas.12422

Franzen, A., & Heinegård, D. (1985). Isolation and characterization of two sialoproteins present only in bone calcified matrix. Biochem J, 232(3), 715-724. doi:10.1042/bj2320715

Gao, S. G., Li, K. H., Zeng, K. B., Tu, M., Xu, M., & Lei, G. H. (2010). Elevated osteopontin level of synovial fluid and articular cartilage is associated with disease severity in knee osteoarthritis patients. Osteoarthritis & Cartilage, 18(1), 82-87. doi:10.1016/j.joca.2009.07.009

Ge, Q., Ruan, C. C., Ma, Y., Tang, X. F., Wu, Q. H., Wang, J. G., . . . Gao, P. J. (2017). Osteopontin regulates macrophage activation and osteoclast formation in hypertensive patients with vascular calcification. Scientific Reports, 7, 40253. doi:10.1038/srep40253

Georgiev, T., & Angelov, A. K. (2019). Modifiable risk factors in knee osteoarthritis: Treatment implications. Rheumatology International, 39(7), 1145-1157. doi:10.1007/s00296-019-04290-z

Gericke, A., Qin, C., Spevak, L., Fujimoto, Y., Butler, W. T., Sørensen, E. S., & Boskey, A. L. (2005). Importance of phosphorylation for osteopontin regulation of biomineralization. Calcified Tissue International, 77(1), 45-54. doi:10.1007/s00223-004-1286-1

Glyn-Jones, S., Palmer, A. J. R., Agricola, R., Price, A. J., Vincent, T. L., Weinans, H., & Carr, A. J. (2015). Osteoarthritis. The Lancet, 386(9991), 376-387. doi:10.1016/S0140-6736(14)60802-3

Goldberg, H., Warner, K., Li, M., & Hunter, G. (2001). Binding of bone sialoprotein, osteopontin and synthetic polypeptides to hydroxyapatite. Connective Tissue Research, 42(1):25-37. doi:10.3109/03008200109014246

Gramoun, A., Goto, T., Nordström, T., Rotstein, O. D., Grinstein, S., Heersche, J. N. M., & Manolson, M. F. (2010). Bone matrix proteins and extracellular acidification: Potential co-regulators of osteoclast morphology. Journal of Cellular Biochemistry, 111(2), 350-361. doi:10.1002/jcb22705

Gray, H.,. (1918). Anatomy of the human body, by Henry Gray. 20th ed., thoroughly rev. and re-edited by Warren H. Lewis. Lea&Febiger, Philadelphia, USA

Page 90: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

88

Greisen, S. R., Einarsson, H. B., Hvid, M., Hauge, E., Deleuran, B., & Kragstrup, T. W. (2015). Spontaneous generation of functional osteoclasts from synovial fluid mononuclear cells as a model of inflammatory osteoclastogenesis. Apmis, 123(9), 779-786. doi:10.1111/apm.12416

Hadhidakis, D. J., & Androulakis, I. I. (2006). Bone remodeling. Annals of the New York Academy of Sciences, 1092(1), 385-396. doi:10.1196/annals.1365.035

Halleen, J. M., Ylipahkala, H., Alatalo, S. L., Janckila, A. J., Heikkinen, J. E., Suominen, H., . . . Väänänen, H. K. (2002). Serum tartrate-resistant acid phosphatase 5b, but not 5a, correlates with other markers of bone turnover and bone mineral density. Calcified Tissue International, 71(1), 20-25. doi:10.1007/s00223-001-2122-7

Halleen, J., Tiitinen, S., Ylipahkala, H., Fagerlund, K., & Väänänen, H. (2006) Tartrate-resistant acid phosphatase 5b (TRACP 5b) as a marker of bone resorption. Clinical Laboratory, 52(9-10):499-509.

Halling Linder, C., Ek-Rylander, B., Krumpel, M., Nordgård, M., Narisawa, S., Millán, J. L., . . . Magnusson, P. (2017). Bone alkaline phosphatase and tartrate-resistant acid phosphatase: Potential co-regulators of bone mineralization. Calcified Tissue International, 101(1), 92-101. doi:10.1007/s00223-017-0259-2

Hampel, U., Sesselmann, S., Iserovich, P., Sel, S., Paulsen, F., & Sack, R. (2013). Chemokine and cytokine levels in osteoarthritis and rheumatoid arthritis synovial fluid. Journal of Immunological Methods, 396(1-2), 134-139. doi:10.1016/j.jim.2013.08.007

Harre, U., Lang, S. C., Pfeifle, R., Rombouts, Y., Frühbeißer, S., Amara, K., . . . Schett, G. (2015). Glycosylation of immunoglobulin G determines osteoclast differentiation and bone loss. Nature Communications, 6, 6651. doi:10.1038/ncomms7651

Hasegawa, M., Nakoshi, Y., Iino, T., Sudo, A., Segawa, T., Maeda, M., . . . Uchida, A. (2009). Thrombin-cleaved osteopontin in synovial fluid of subjects with rheumatoid arthritis. Journal of Rheumatology, 36(2), 240-245. doi:10.3899/jrheum.080753

Hayman, A. R., Jones, S. J., Boyde, A., Foster, D., Colledge, W. H., Carlton, M. B., . . . Cox, T. M. (1996). Mice lacking tartrate-resistant acid phosphatase (ACP5) have disrupted endochondral ossification and mild osteopetrosis. Development 122(10), 3151-62.

He, B., Wu, J. P., Kirk, T. B., Carrino, J. A., Xiang, C., & Xu, J. (2014). High-resolution measurements of the multilayer ultra-structure of articular cartilage and their translational potential. Arthritis Research & Therapy, 16(2), 205. doi:10.1186/ar4506

Hemingway, F., Cheng, X., Knowles, H. J., Estrada, F. M., Gordon, S., & Athanasou, N. A. (2011). In vitro generation of mature human osteoclasts. Calcified Tissue International, 89(5), 389-395. doi:10.1007/s00223-011-9530-0

Henriksen, K., Sørensen, M.,G., Nielsen, R. H., Gram, J., Schaller, S., Dziegiel, M. H., . . . Karsdal, M. A. (2006). Degradation of the organic phase of bone by osteoclasts: A secondary role for lysosomal acidification. Journal of Bone and Mineral Research, 21(1), 58-66. doi:10.1359/JBMR.050905

Hoac, B., Nelea, V., Jiang, W., Kaartinen, M. T., & McKee, M. D. (2017). Mineralization-inhibiting effects of transglutaminase-crosslinked polymeric osteopontin. Bone, 101, 37-48. doi:10.1016/j.bone.2017.04.007

Page 91: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

89

Holm, E., Gleberzon, J. S., Liao, Y., Sørensen, E. S., Beier, F., Hunter, G. K., & Goldberg, H. A. (2014). Osteopontin mediates mineralization and not osteogenic cell development in vitro. Biochemical Journal, 464(3), 355-364. doi:10.1042/BJ20140702

Honsawek, S., Tanavalee, A., Sakdinakiattikoon, M., Chayanupatkul, M., & Yuktanandana, P. (2009). Correlation of plasma and synovial fluid osteopontin with disease severity in knee osteoarthritis. Clinical Biochemistry, 42(9), 808-812. doi:10.1016/j.clinbiochem.2009.02.002

Horton, J. E., Raisz, L. G., Simmons, H. A., Oppenheim, J. J., & Mergenhagen, S. E. (1972). Bone resorbing activity in supernatant fluid from cultured human peripheral blood leukocytes. Science, 177(4051), 793. doi:10.1126/science.177.4051.793

Hou, W. S., Li, W., Keyszer, G., Weber, E., Levy, R., Klein, M. J., . . . Brömme, D. (2002). Comparison of cathepsins K and S expression within the rheumatoid and osteoarthritic synovium. Arthritis & Rheumatism, 46(3), 663-674. doi:10.1002/art.10114

Huang, Y. J., Huang, T. W., Chao, T. Y., Sun, Y. S., Chen, S. J., Chu, D. M., . . . Wu, L. W. (2017). Elevated serum tartrate-resistant acid phosphatase isoform 5a levels in metabolic syndrome. Oncotarget 8(44), 78144-78152. doi:10.18632/oncotarget.17839

Hügle, T., & Geurts, J. (2017). What drives osteoarthritis? -synovial versus subchondral bone pathology. Rheumatology, 56(9), 1461-1471. doi:10.1093/rheumatology/kew389

Hussein, W. M., Feder, D., Schenk, G., Guddat, L. W., & McGeary, R. P.. (2018). Synthesis and evaluation of novel purple acid phosphatase inhibitors. Medicinal Chemistry Communication 10(1), 61-761. doi:10.1039/C8MD00491A

Icer, M. A., & Gezmen-Karadag, M. (2018). The multiple functions and mechanisms of osteopontin. Clinical Biochemistry, 59, 17-24. doi:.1016/j.clinbiochem.2018.07.003

Im, G., & Kim, M. (2014). The relationship between osteoarthritis and osteoporosis. Journal of Bone and Mineral Metabolism, 32(2), 101-109. doi:10.1007/s00774-013-0531-0

Iwadate, H., Kobayashi, H., Kanno, T., Asano, T., Saito, R., Sato, S., . . . Ohira, H. (2013). Plasma osteopontin is correlated with bone resorption markers in rheumatoid arthritis patients. International Journal of Rheumatic Diseases, 17(1), 50-56. doi:10.1111/1756-185X.12115

Iwanaga, T., Shikichi, M., Kitamura, H., Yanase, H., & Nozawa-Inoue, K. (2000). Morphology and functional roles of synoviocytes in the joint. Archives of Histology and Cytology, 63(1), 17-31. doi:10.1679/aohc.63.17

Izumi, K., Kaneko, Y., Hashizume, M., Yoshimoto, K., & Takeuchi, T. (2015). Baseline serum osteopontin levels predict the clinical effectiveness of tocilizumab but not infliximab in biologic-naive patients with rheumatoid arthritis: A single-center prospective study at 1 year (the Keio first-bio cohort study). PLoS One, 10(12), e0145468. doi:10.1371/journal.pone.0145468

Janckila, A. J., & Yam, L. T. (2009). Biology and clinical significance of tartrate-resistant acid phosphatases: New perspectives on an old enzyme. Calcified Tissue International, 85(6), 465-483. doi:10.1007/s00223-009-9309-8

Janckila, A. J., Lederer, E. D., Price, B. A., & Yam, L. T. (2009). Tartrate-resistant acid phosphatase isoform 5a as an inflammation marker in end-stage renal disease. Clinical nephrology 71(4), 387-396. doi:10.5414/CNP71387

Page 92: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

90

Janckila, A. J., Neustadt, D. H., & Yam, L. T. (2008). Significance of serum TRACP in rheumatoid arthritis. Journal of Bone & Mineral Research, 23(8), 1287-1295. doi:10.1359/jbmr.080329

Janckila, A. J., Takahashi, K., Sun, S. Z., & Yam, L. T. (2001a). Tartrate-resistant acid phosphatase isoform 5b as serum marker for osteoclastic activity. Clinical Chemistry, 47(1), 74-80.

Kariya, Y., Kanno, M., Matsumoto-Morita, K., Konno, M., Yamaguchi, Y., & Hashimoto, Y. (2014). Osteopontin O-glycosylation contributes to its phosphorylation and cell-adhesion properties. Biochemical Journal, 463(1), 93-102. doi:10.1042/BJ20140060

Katsimbri, P. (2017). The biology of normal bone remodelling. European Journal of Cancer Care, 26(6), e12740. doi:10.1111/ecc.12740

Kazanecki, C. C., Uzwiak, D. J., & Denhardt, D. T. (2007). Control of osteopontin signaling and function by post-translational phosphorylation and protein folding. Journal of Cellular Biochemistry, 102(4), 912-924. doi:10.1002/jcb.21558

Kellgren, J. H., & Lawrence, J. S. (1957). Radiological assessment of osteo-arthrosis. Annals of the Rheumatic Diseases, 16(4), 494-502. doi:10.1136/ard.16.4.494

Kenkre, J. S., & Bassett, J. (2018). The bone remodelling cycle. Annals of Clinical Biochemistry, 55(3), 308-327. doi:10.1177/0004563218759371

Kierszenbaum, A. L., & Tres, L. L. (2015). Histology and cell biology: An introduction to pathology. 4th edition. Philadelphia, PA: Elsevier Saunders.

Kim, H., Kim, K., Kim, B., Cho, M., & Lee, S. (2015). The effect of vascular endothelial growth factor on osteoclastogenesis in rheumatoid arthritis. PLoS One, 10(4), e0124909. doi:10.1371/journal.pone.0124909

Kim, K. W., Kim, B. M., Moon, H. W, Lee, S. H., & Kim, H. R. (2015). Role of C-reactive protein in osteoclastogenesis in rheumatoid arthritis. Arthritis Research & Therapy, 17(1), 41. doi:10.1186/s13075-015-0563-z

Knee and Hip Osteoarthritis. Current Care Guidelines, 2018. Working group set up by the Finnish Medical Society Duodecim and the Finnish Orthopaedic Association. Helsinki: The Finnish Medical Society Duodecim, 2018 (referred March 30, 2019). Available online at: www.kaypahoito.fi.

Kode, A., Manavalan, J. S., Mosialou, I., Bhagat, G., Rathinam, C. V., Luo, N., . . . Kousteni, S. (2014). Leukaemogenesis induced by an activating β-catenin mutation in osteoblasts. Nature, 506(7487), 240-244. doi:10.1038/nature12883

Kohri, K., Yasui, T., Okada, A., Hirose, M., Hamamoto, S., Fujii, Y., . . . Taguchi, K. (2012). Biomolecular mechanism of urinary stone formation involving osteopontin. Urological Research, 40(6), 623-637. doi:10.1007/s00240-012-0514-y

Kong, Y., Yoshida, H., Sarosi, I., Tan, H., Timms, E., Capparelli, C., . . . Penninger, J. M. (1999). OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature, 397(6717), 315-323. doi:10.1038/16852

Kononoff, A., Arstila, L., Pussinen, P., Kautiainen, H., Elfving, P., Savolainen, E., . . . Kaipiainen-Seppänen, O. (2017). Incidence of inflammatory joint diseases in finland: Results from a population-based epidemiological study. Rheumatology International, 37(10), 1693-1700. doi:10.1007/s00296-017-3779-1

Page 93: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

91

Kotani, M., Kikuta, J., Klauschen, F., Chino, T., Kobayashi, Y., Yasuda, H., . . . Ishii, M. (2013). Systemic circulation and bone recruitment of osteoclast precursors tracked by using fluorescent imaging techniques. The Journal of Immunology, 190(2), 605-612. doi:10.4049/jimmunol.1201345

Krishnamurthy, A., Joshua, V., Haj Hensvold, A., Jin, T., Sun, M., Vivar, N., . . . Catrina, A. I. (2016). Identification of a novel chemokine-dependent molecular mechanism underlying rheumatoid arthritis-associated autoantibody-mediated bone loss. Annals of the Rheumatic Diseases, 75(4), 721-729. doi:10.1136/annrheumdis-2015-208093

Krum, S. A., Miranda-Carboni, G., Hauschka, P. V., Carroll, J. S., Lane, T. F., Freedman, L. P., & Brown, M. (2008). Estrogen protects bone by inducing fas ligand in osteoblasts to regulate osteoclast survival. The EMBO Journal, 27(3), 535-545. doi:10.1038/sj.emboj.7601984

Kurko, J., Besenyei, T., Laki, J., Glant, T. T., Mikecz, K., & Szekanecz, Z. (2013). Genetics of rheumatoid arthritis - A comprehensive review. Clinical Reviews in Allergy & Immunology, 45(2), 170-179. doi:10.1007/s12016-012-8346-7

Kurowska-Stolarska, M., & Alivernini, S. (2017). Synovial tissue macrophages: Friend or foe? RMD Open, 3(2), e000527. doi:10.1136/rmdopen-2017-000527

Kusuyama, J., Bandow, K., Ohnishi, T., Hisadome, M., Shima, K., Semba, I., & Matsuguchi, T. (2017). Osteopontin inhibits osteoblast responsiveness through the down-regulation of focal adhesion kinase mediated by the induction of lower molecular weight protein tyrosine phosphatase. Molecular Biology of the Cell, 28(10), 1326-1336. doi:10.1091/mbc.E16-10-0716

Lai, X., Wang, H., Cao, J., Li, Y., Dai, Y., Xiang, Y., & Zhang, L. (2016). Circulating IL-27 is elevated in rheumatoid arthritis patients Molecules, 21(11), e1565. doi:10.3390/molecules21111565

Lång, P., & Andersson, G. (2005). Differential expression of monomeric and proteolytically processed forms of tartrate-resistant acid phosphatase in rat tissues. Cellular and Molecular Life Sciences, 62(7-8), 905-918. doi:10.1007/s00018-005-4550-4

Li, H., Cui, Y., Luan, J., Zhang, X., Li, C., Zhou, X., . . . Han, J. (2016). PRELP (proline/arginine-rich end leucine-rich repeat protein) promotes osteoblastic differentiation of preosteoblastic MC3T3-E1 cells by regulating the β-catenin pathway. Biochemical and Biophysical Research Communications 470(3), 558-562. doi:1016/j.bbrc.2016.01.106

Li, H., Lu, Y., Qian, J., Zheng, Y., Zhang, M., Bi, E., . . . Yi, Q. (2014). Human osteoclasts are inducible immunosuppressive cells in response to T cell derived IFN- and CD40 ligand in vitro. Journal of Bone and Mineral Research, 29(12), 2666-2675. doi:10.1002/jbmr.2294

Li, H., Shen, H., Yan, G., Zhang, Y., Liu, M., Fang, P., . . . Yang, P. (2015). Site-specific structural characterization of O-glycosylation and identification of phosphorylation sites of recombinant osteopontin. Biochimica et Biophysica Acta (BBA) - Proteins and Proteomics, 1854(6), 581-591. doi:10.1016/j.bbapap.2014.09.025.

Page 94: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

92

Li, J., Sarosi, I., Yan, X. Q., Morony, S., Capparelli, C., Tan, H. L., . . . Boyle, W. J. (2000). RANK is the intrinsic hematopoietic cell surface receptor that controls osteoclastogenesis and regulation of bone mass and calcium metabolism. Proceedings of the National Academy of Sciences USA, 97(4), 1566-1571. doi:10.1073/pnas.97.4.1566

Listing, J., Kekow, J., Manger, B., Burmester, G., Pattloch, D., Zink, A., & Strangfeld, A. (2015). Mortality in rheumatoid arthritis: The impact of disease activity, treatment with glucocorticoids, TNF- inhibitors and rituximab. Annals of the Rheumatic Diseses, 74(2), 415421. doi:10.1136/annrheumdis-2013-204021

Ljusberg, J., Wang, Y., Lång, P., Norgård, M., Dodds, R., Hultenby, K., . . . Andersson, G. (2005). Proteolytic excision of a repressive loop domain in tartrate-resistant acid phosphatase by cathepsin K in osteoclasts. Journal of Biological Chemistry, 280(31), 28370-28381. doi:10.1074/jbc.M502469200

Lotinun, S., Kiviranta, R., Matsubara, T., Alzate, J. A., Neff, L., Lüth, A., . . . Baron, R. (2013). Osteoclast-specific cathepsin K deletion stimulates S1P-dependent bone formation. Journal of Clinical Investigations, 123(2), 666-681. doi:10.1172/JCI64840

Lotz, M., Martel-Pelletier, J., Christiansen, C., Brandi, M. L., Bruyère, O., Chapurlat, R., . . . Reginster, J. Y. (2013). Value of biomarkers in osteoarthritis: Current status and perspectives. Annals of the Rheumatic Diseases, 72(11), 1756-1763. doi:10.1136/annrheumdis-2013-203726

Lowell, C. A., Niwa, M., Soriano, P., & Varmus, H. E. (1996). Deficiency of the hck and src tyrosine kinases results in extreme levels of extramedullary hematopoiesis. Blood, 87(5), 1780-1792.

Lund, S. A., Giachelli, C. M., & Scatena, M. (2009). The role of osteopontin in inflammatory processes. Journal of Cell Communication and Signaling, 3(3-4), 311-322. doi:10.1007/s12079-009-0068-0

Maeda, H., Kukita, T., Akamine, A., Kukita, A., & Iijima, T. (1994). Localization of osteopontin in resorption lacunae formed by osteoclast-like cells: A study by a novel monoclonal antibody which recognizes rat osteopontin. Histochemistry, 102(4), 247-254.

Mansour, A., Mezour, M. A., Badran, Z., & Tamimi, F. (2017). Extracellular matrices for bone regeneration: A literature review. Tissue Engineering. Part A, 23(23-24), 1436-1451. doi:10.1089/ten.TEA.2017.0026

Mathiessen, A., & Conaghan, P. G. (2017). Synovitis in osteoarthritis: Current understanding with therapeutic implications. Arthritis Research & Therapy 19(1), 18. doi:10.1186/s13075-017-1229-9

Matsui, Y., Iwasaki, N., Kon, S., Takahashi, D., Morimoto, J., Matsui, Y., . . . Uede, T. (2009). Accelerated development of aging-associated and instability-induced osteoarthritis in osteopontin-deficient mice. Arthritis & Rheumatism, 60(8), 2362-2371. doi:10.1002/art.24705

Matsuo, K., & Irie, N. (2008). Osteoclast–osteoblast communication. Archives of Biochemistry and Biophysics 473(2), 201-209. doi:10.1016/j.abb.2008.03.027

Page 95: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

93

McInnes, I. B., & Schett, G. (2011). The pathogenesis of rheumatoid arthritis. New England Journal of Medicine, 365(23), 2205-2219. doi:10.1056/NEJMra1004965

Mizoguchi, T., Muto, A., Udagawa, N., Arai, A., Yamashita, T., Hosoya, A., . . . Takahashi, N. (2009). Identification of cell cycle arrested quiescent osteoclast precursors in vivo. Journal of Cell Biology, 184(4), 541-554. doi:10.1083/jcb.200806139

Monasterio, G., Castillo, F., Rojas, L., Cafferata, E. A., Alvarez, C., Carvajal, P., . . . Vernal, R. (2018). Th1/Th17/Th22 immune response and their association with joint pain, imagenological bone loss, RANKL expression and osteoclast activity in temporomandibular joint osteoarthritis: A preliminary report. Journal of Oral Rehabilitation, 45(8), 589-597. doi:10.1111/joor.12649

Morgan, S., Poundarik, A. A., & Vashishth, D. (2015). Do non-collagenous proteins affect skeletal mechanical properties? Calcified Tissue International, 97(3), 281-291. doi:10.1007/s00223-015-0016-3

Morimoto, J., Kon, S., Matsui, Y., & Uede, T. (2010). Osteopontin; as a target molecule for the treatment of inflammatory diseases. Current Drug Targets, 11(4), 494-505. doi:10.2174/138945010790980321

Moser, S. C., & van, der Erden, B. C. J. (2019). Osteocalcin - A versatile bone-derived hormone. Frontiers in Endocrinology, 9, 794. doi:10.3389/fendo.2018.00794

Nakamura, M., Hentunen, T., Salonen, J., Nagai, A., & Yamashita, K. (2013). Characterization of bone mineral-resembling biomaterials for optimizing human osteoclast differentiation and resorption. Journal of Biomedical Materials Research Part A, 101(11), 3141-3151. doi:10.1002/jbm.a.34621

Nau, G. J., Liaw, L., Chupp, G. L., Berman, J. S., Hogan, B. L. M., & Young, R. A. (1999). Attenuated host resistance against mycobacterium bovis BCG infection in mice lacking osteopontin. Infection and Immunity, 67(8), 4223-4230.

Nikiphorou, E., Konan, S., MacGregor, A. J., Haddad, F. S., & Young, A. (2014). The surgical treatment of rheumatoid arthritis. The Bone & Joint Journal, 96-B(10), 1287-1289. doi:10.1302/0301-620X.96B10.34506

Nyström, T., Dunér, P., & Hultgårdh-Nilsson, A. (2007). A constitutive endogenous osteopontin production is important for macrophage function and differentiation. Experimental Cell Research, 313(6), 1149-1160. doi:10.1016/j.yexcr.2006.12.026

Okamoto, K., & Takayanagi, H. (2019). Osteoimmunology. Cold Spring Harbor Perspectives in Medicine, 9(1). doi:10.1016/B978-0-12-804182-6.00016-2

Orlowsky, E. W., & Kraus, V. B. (2015). The role of innate immunity in osteoarthritis: When our first line of defense goes on the offensive. Journal of Rheumatology, 42(3), 363-371. doi:10.3899/jrheum.140382

Ouyang, W., Kolls, J. K., & Zheng, Y. (2008). The biological functions of T helper 17 cell effector cytokines in inflammation. Immunity, 28(4), 454-467. doi:10.1016/j.immuni.2008.03.004

Paiva, K. B. S., & Granjeiro, J. M. (2017). Chapter six - matrix metalloproteinases in bone resorption, remodeling, and repair. In R. A. Khalil (Ed.), Progress in molecular biology and translational science (pp. 203-303) Academic Press. doi:10.1016/bs.pmbts.2017.05.001

Page 96: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

94

Palazzo, C., Nguyen, C., Lefevre-Colau, M., Rannou, F., & Poiraudeau, S. (2016). Risk factors and burden of osteoarthritis. Annals of Physical and Rehabilitation Medicine, 59(3), 134-138. doi:10.1016/j.rehab.2016.01.006

Pappalardo, A., & Thompson, K. (2015). Novel immunostimulatory effects of osteoclasts and macrophages on human ϒδ T cells. Bone, 71, 180-188. doi:10.1016/j.bone.2014.10.019

Park-Min, K. H. (2018). Mechanisms involved in normal and pathological osteoclastogenesis. Cellular and Molecular Life Sciences, 75(14), 2519-2528. doi:10.1007/s00018-018-2817-9

Pavlovic, V., Dimic, A., Milenkovic, S., & Krtinic. (2014). Serum levels of IL-17, IL-4, and INFgamma in serbian patients with early rheumatoid arthritis. Journal of Research in Medical Sciences 19(1), 18–22. doi:10.1515/med-2015-0019

Pettit, A. R., Ji, H., von Stechow, D., Müller, R., Goldring, S. R., Choi, Y., . . . Gravallese, E. M. (2001). TRANCE/RANKL knockout mice are protected from bone erosion in a serum transfer model of arthritis. The American Journal of Pathology, 159(5), 1689-1699. doi:10.1016/S0002-9440(10)63016-7

Pettit, A. R., Walsh, N. C., Manning, C., Goldring, S. R., & Gravallese, E. M. (2006). RANKL protein is expressed at the pannus-bone interface at sites of articular bone erosion in rheumatoid arthritis. Rheumatology, 45(9), 1068-1076. doi:10.1093/rheumatology/kel045

Piao, Z., & Yuan, H. (2017). Osteopontin exacerbates pseudomonas aeruginosa-induced bacteremia in mice. Cellular Immunology, 318, 23-28. doi:10.1016/j.cellimm.2017.05.004

Pierce, A.M., Lindskog, S., Hammarström, L. (1991) Osteoclasts: Structure and function. Electron Microscopy Reviews, 4(1):1-45. doi:10.1016/0892-0354(91)90015-5

Pietras, A., Katz, A. M., Ekström, E.,J., Wee, B., Halliday, J. J., Pitter, K. L., . . . Holland, E. C. (2014). Osteopontin-CD44 signaling in the glioma perivascular niche enhances cancer stem cell phenotypes and promotes aggressive tumor growth. Cell Stem Cell, 14(3), 357-369. doi:10.1016/j.stem.2014.01.005

Pilichou, A., Papassotiriou, I., Michalakakou, K., Fessatou, S., Fandridis, E., Papachristou, G., & Terpos, E. (2008). High levels of synovial fluid osteoprotegerin (OPG) and increased serum ratio of receptor activator of nuclear factor-ΚB ligand (RANKL) to OPG correlate with disease severity in patients with primary knee osteoarthritis. Clinical biochemistry 41(9), 746-749. doi:10.1016/j.clinbiochem.2008.02.011

Pozgan, U., Caglic, D., Rozman, B., Nagase, H., Turk, V., & Turk, B. (2010). Expression and activity profiling of selected cysteine cathepsins and matrix metalloproteinases in synovial fluids from patients with rheumatoid arthritis and osteoarthritis. Biological Chemistry, 391(5), 571-579. doi:10.1515/BC.2010.035

Prieto-Potin, I., Largo, R., Roman-Blas, J., Herrero-Beaumont, G., & Walsh, D. A. (2015). Characterization of multinucleated giant cells in synovium and subchondral bone in knee osteoarthritis and rheumatoid arthritis. BMC Musculoskeletal Disorders, 16, 226. doi:10.1186/s12891-015-0664-5

Page 97: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

95

Qin, C., Baba, O., & Butler, W. T. (2004). Post-translational modifications of SIBLING proteins and their roles in osteogenesis and dentinogenesis. Critical Reviews in Oral Biology & Medicine, 15(3), 126-136. doi:10.1177/154411130401500302

Repp, F., Kollmannsberger, P., Roschger, A., Berzlanovich, A., Gruber, G. M., Roschger, P., . . . Weinkamer, R. (2017). Coalignment of osteocyte canaliculi and collagen fibers in human osteonal bone. Journal of Structural Biology 199(3), 177-186. doi:10.1016/j.jsb.2017.07.004.

Rheumatoid Arthritis. Current Care Guidelines, 2015. Working group set up by The Finnish Medical Society Duodecim and The Finnish Society for Rheumatology. Helsinki: The Finnish medical Society Duodecim, 2015 (referred march 23, 2019). Available online at: www.kaypahoito.fi.

Robling, A. G., & Turner, C. H. (2009). Mechanical signaling for bone modeling and remodeling. Critical reviews in eukaryotic gene expression, 19(4), 319–338. doi:10.1615/CritRevEukarGeneExpr.v19.i4.50

Rodan, G. A., & Martin, T. J. (2000). Therapeutic approaches to bone diseases. Science, 289(5484), 1508-1514. doi:10.1126/science.289.5484.1508

Roeder, E., Matthews, B. G., & Kalajzic, I. (2016). Visual reporters for study of the osteoblast lineage. Bone 92, 189-195. doi:10.1016/j.bone.2016.09.004

Ross, F. P., Chappel, J., Alvarez, J. I., Sander, D., Butler, W. T., Farach-Carson, M., . . . Cheresh, D. A. (1993). Interactions between the bone matrix proteins osteopontin and bone sialoprotein and the osteoclast integrin alpha v beta 3 potentiate bone resorption. Journal of Biological Chemistry, 268(13), 9901-9907.

Rosset, E. M., & Bradshaw, A. D. (2016). SPARC/osteonectin in mineralized tissue. Matrix Biology, 52-54, 78-87 doi:10.1016/j.matbio.2016.02.001

Rucci, N., & Teti, A. (2016). The “love–hate” relationship between osteoclasts and bone matrix. Matrix Biology 52-54, 176-190. doi:10.1016/j.matbio.2016.02.009

Sainger, R., Grau, J. B., Poggio, P., Branchetti, E., Bavaria, J. E., Gorman, J. H. 3rd, . . . Ferrari, G. (2012). Dephosphorylation of circulating human osteopontin correlates with severe valvular calcification in patients with calcific aortic valve disease. Biomarkers, 17(2), 111-118. doi:10.3109/1354750X.2011.642407

Sato, M., Asada, N., Kawano, Y., Wakahashi, K., Minagawa, K., Kawano, H., . . . Katayama, Y. (2013). Osteocytes regulate primary lymphoid organs and fat metabolism. Cell Metabolism 18(5), 749-758. doi:10.1016/j.cmet.2013.09.014

Schett, G., & Gravallese, E. (2012). Bone erosion in rheumatoid arthritis: Mechanisms, diagnosis and treatment. Nature Reviews in Rheumatology, 8(11), 656-664. doi:10.1038/nrrheum.2012.153

Scublinsky, D., & Gonzalez, C. D. (2016). Quantifying disease in challenging conditions: Incidence and prevalence of rheumatoid arthritis. Journal of Rheumatology, 43(7), 1263-1264. doi:10.3899/jrheum.160522

Seeman, E., & Delmas, P. D. (2006). Bone quality − the material and structural basis of bone strength and fragility. New England Journal of Medicine, 354(21), 2250-2261. doi:10.1056/NEJMra053077

Page 98: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

96

Selander, K., Lehenkari, P., & Väänänen, H. K. (1994). The effects of bisphosphonates on the resorption cycle of isolated osteoclasts. Calcified Tissue International, 55(5), 368-375. doi:10.1007/BF00299317

Sellam, J., Rivière, E., Courties, A., Rouzaire, P-O.., Tolusso, B., Vital, E. M., . . . Mariette, X. (2016). Serum IL-33, a new marker predicting response to rituximab in rheumatoid arthritis. Arthritis Research & Therapy, 18(1), 294. doi:10.1186/s13075-016-1190-z

Seol, J. W., Lee, H. B., Kim, N. S., & Park, S. Y. (2009). Tartrate-resistant acid phosphatase as a diagnostic factor for arthritis. International Journal of Molecular Medicine, 24(1), 57-62. doi:10.3892/ijmm_00000206

Shim, J. H., Stavre, Z., & Gravallese, E. M. (2018). Bone loss in rheumatoid arthritis: Basic mechanisms and clinical implications. Calcified Tissue International, 102(5), 533-546. doi:10.1007/s00223-017-0373-1

Shimazu, Y., Nanci, A., & Aoba, T. (2002). Immunodetection of osteopontin at sites of resorption in the pulp of rat molars. Journal of Histochemistry & Cytochemistry, 50(7), 911-921. doi:10.1177/0022125540205000705

Shio, K., Kobayashi, H., Asano, T., Saito, R., Iwadate, H., Watanabe, H., . . . Ohira, H. (2010). Thrombin-cleaved osteopontin is increased in urine of patients with rheumatoid arthritis. Journal of Rheumatology, 37(4), 704-710. doi:10.3899/jrehum090582

Silva, B. C., & Bilezikian, J. P. (2015). Parathyroid hormone: Anabolic and catabolic actions on the skeleton. Current Opinion in Pharmacology 22, 41-50. doi:10.1016/j.coph.2015.03.005

Simonet, W. S., Lacey, D. L., Dunstan, C. R., Kelley, M., Chang, M. S., Lüthy, R., . . . Boyle, W. J. (1997). Osteoprotegerin: A novel secreted protein involved in the regulation of bone density. Cell, 89(2), 309-319. doi:10.1016/S0092-8674(00)80209-3

Skoumal, M., Haberhauer, G., Kolarz, G., Hawa, G., Woloszczuk, W., & Klingler, A. (2004). Serum cathepsin K levels of patients with longstanding rheumatoid arthritis: Correlation with radiological destruction. Arthritis Research & Therapy, 7(1), R65-70. doi:10.1186/ar1461

Smith M. D.. The normal synovium. The Open Rheumatology Journal, 5, 100–106. doi:10.2174/1874312901105010100

Smolen, J. S., Aletaha, D., & McInnes, I. B. (2016). Rheumatoid arthritis. The Lancet, 388(10055), 2023-2038. doi:10.1016/S0140-6736(16)30173-8

Smolen, J. S., Landewé, R., Bijlsma, J., Burmester, G., Chatzidionysiou, K., Dougados, M., . . . van der Heijde, D. (2014). EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update. Annals of the Rheumatic Diseases, 76(6), 960-977. doi:10.1136/annrheumdis-2016-210715

Sodek, J., Ganss, B., & McKee, M. D. (2000). Osteopontin. Critical Reviews in Oral Biology & Medicine, 11(3), 279-303. doi:10.1177/10454411000110030101

Sørensen, M. G., Henriksen, K., Schaller, S., Henriksen, D. B., Nielsen, F. C., Dziegiel, M. H., & Karsdal, M. A. (2007). Characterization of osteoclasts derived from CD14+ monocytes isolated from peripheral blood. Journal of Bone and Mineral Metabolism, 25(1), 36-45. doi:10.1007/s00774-006-0725-9

Page 99: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

97

Soysa, N. S., & Alles, N. (2016). Osteoclast function and bone-resorbing activity: An overview. Biochemical and Biophysical Research Communications, 476(3):115-120. doi:10.1016/j.bbrc.2016.05.019

Stock, S. R. (2015). The Mineral-Collagen interface in bone. Calcified Tissue International, 97(3), 262-280. doi:10.1007/s00223-015-9984-6

Sulzbacher, I. (2013). Osteoarthritis: Histology and pathogenesis. Wiener Medizinische Wochenschrift, 163(9-10), 212-219. doi:10.1007/s10354-012-0168-y

Suzuki, Y., Tsutsumi, Y., Nakagawa, M., Suzuki, H., Matsushita, K., Beppu, M., . . . Mizushima, Y. (2001). Osteoclast-like cells in an in vitro model of bone destruction by rheumatoid synovium. Rheumatology, 40(6), 673-682. doi:10.1093/rheumatology/40.6.673

Tagliabracci, V. S., Engel, J. L., Wen, J., Wiley, S. E., Worby, C. A., Kinch, L. N., . . . Dixon, J. E. (2012). Secreted kinase phosphorylates extracellular proteins that regulate biomineralization. Science, 336(6085), 1150. doi:10.1126/science.1217817

Tai, T. W., Chen, C. Y., Su, F. C., Tu, Y. K., Tsai, T. T, Lin, C. F., & Jou, I. M. (2017). Reactive oxygen species are required for zoledronic acid-induced apoptosis in osteoclast precursors and mature osteoclast-like cells. Scientific Reports, 7, 44245. doi:10.1038/srep44245

Takahashi, N., Udagawa, N., & Suda, T. (2014). Vitamin D endocrine system and osteoclasts. BoneKEy Reports, 5(3), 495. doi:10.1038/bonekey.2013.229

Takayanagi, H., Ogasawara, K., Hida, S., Chiba, T., Murata, S., Sato, K., . . . Taniguchi, T. (2000). T-cell-mediated regulation of osteoclastogenesis by signalling cross-talk between RANKL and IFN-gamma. Nature, 408, 600-605. doi:10.1038/35046102

Takito, J., Inoue, S., & Nakamura, M. (2018). The sealing zone in osteoclasts: A self-organized structure on the bone. International Journal of Moleculelar Science, 19(4), E984. doi:10.3390/ijms19040984

Teitelbaum, S. L. (2007). Osteoclasts: What do they do and how do they do it? American Journal of Pathology, 170(2), 427-35. doi:10.2353/ajpath.2007.060834

Terashima, A., & Takayanagi, H. (2018). Overview of osteoimmunology. Calcified Tissue International, 102(5), 503-511. doi:10.1007/s00223-018-0417-1

Teti, A. (2013). Mechanisms of osteoclast-dependent bone formation. BoneKEy Reports, 2, 449. doi:10.1038/bonekey.2013.183

Trieb, K., & Hofstaetter, S. G. (2009). Treatment strategies in surgery for rheumatoid arthritis. European Journal of Radiology, 71(2), 204-210. doi:10.1016/j.ejrad.2009.04.050

Tsuboi, H., Matsui, Y., Hayashida, K., Yamane, S., Maeda-Tanimura, M., Nampei, A., . . . Ochi, T. (2003). Tartrate resistant acid phosphatase (TRAP) positive cells in rheumatoid synovium may induce the destruction of articular cartilage. Annals of the Rheumatic Diseases, 62(3), 196-203. doi:10.1136/ard.62.3.196

Väänänen, H. K., Zhao, H., Mulari, M., & Halleen, J. M. (2000). The cell biology of osteoclast function. Journal of Cell Science, 113(3), 377-381.

Page 100: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

98

Vangsness, C. T. Jr., Burke, W. S., Narvy, S. J., MacPhee, R. D., & Fedenko, A, N. (2011) Human knee synovial fluid cytokines correlated with grade of knee osteoarthritis – a pilot study. Bulletin of the NYU Hospital for Joint Diseases, 69(2), 122-7.

Wallenius, M., Skomsvoll, J. F., Irgens, L. M., Salvesen, K. Ã., Koldingsnes, W., Mikkelsen, K., . . . Kvien, T. K. (2010). Postpartum onset of rheumatoid arthritis and other chronic arthritides: Results from a patient register linked to a medical birth registry. Annals of the Rheumatic Diseases, 69(2), 332-336. doi:10.1136/ard.2009.115964

Wang, K. X., & Denhardt, D. T. (2008). Osteopontin: Role in immune regulation and stress responses. Cytokine & Growth Factor Reviews, 19(5–6), 333-345. doi:10.1016/j.cytogfr.2008.08.001

Wang, L., Guan, X., Tang, R., Hoyer, J. R., Wierzbicki, A., De Yoreo, J. J., & Nancollas, G. H. (2008). Phosphorylation of osteopontin is required for inhibition of calcium oxalate crystallization. Journal of Physical Chemistry B, Condensed Matter, Materials, Surfaces, Interfaces & Biophysical, 112(30), 9151-9157. doi:10.1021/jp804282u

Weatherholt, A. M., Fuchs, R. k., & Warden, S. J. (2011). Specialized connective tissue: Bone, the structural framework of the upper extremity. Journal of Hand Therapy, 25(2):123-131. doi:10.1016/j.jht.2011.08.003

Weber, G. F., Zawaideh, S., Hikita, S., Kumar, V. A., Cantor, H., & Ashkar, S. (2002). Phosphorylation-dependent interaction of osteopontin with its receptors regulates macrophage migration and activation. Journal of Leukocyte Biology, 72(4), 752-761. doi:10.1189/jlb.72.4.752

Weissmann, G. (2004). Pathogenesis of rheumatoid arthritis. JCR: Journal of Clinical Rheumatology, 10(3) (Supplement), S26-S31. doi:10.1097/01.rhu.0000130687.75646.44

Whyte, M. P., McAlister, W. H., Fallon, M. D., Pierpont, M. E., Bijanki, V. N., Duan, S., . . . Mumm, S. (2017). Raine syndrome (OMIM #259775), caused by FAM20C mutation, is congenital sclerosing osteomalacia with cerebral calcification (OMIM 259660). Journal of Bone and Mineral Research, 32(4), 757-769. doi:10.1002/jbmr.3034

Wojdasiewicz, P., Poniatowski, L. A., & Szukiewicz, D. (2014). The role of inflammatory and anti-inflammatory cytokines in the pathogenesis of osteoarthritis. Mediators of Inflammation, 2014, 561459. doi:10.1155/2014/561459

Woolf, A., & Pfleger, B. (2003). Burden of major musculoskeletal conditions. Bulletin of the World Health Organisation 81(9), 646-656. doi:10.1590/S0042-96862003000900007

Xu, G., Nie, H., Li, N., Zheng, W., Zhang, D., Feng, G., . . . Zhang, J. Z. (2005). Role of osteopontin in amplification and perpetuation of rheumatoid synovitis. The Journal of Clinical Investigation, 115(4), 1060-1067. doi:10.1172/JCI23273

Xu, M., Zhang, L., Zhao, L., Gao, S., Han, R., Su, D., & Lei, G. (2013). Phosphorylation of osteopontin in osteoarthritis degenerative cartilage and its effect on matrix metalloprotease 13. Rheumatology International, 33(5), 1313-1319. doi:10.1007/s00296-012-2548-4

Page 101: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

99

Yamaguchi, R., Yamamoto, T., Motomura, G., Ikemura, S., Iwasaki, K., Zhao, G., . . . Iwamoto, Y. (2014). Bone and cartilage metabolism markers in synovial fluid of the hip joint with secondary osteoarthritis. Rheumatology, 53(12), 2191–2195, doi:10.1093/rheumatology/keu253

Yamamoto, N., Sakai, F., Kon, S., Morimoto, J., Kimura, C., Yamazaki, H., . . . Uede, T. (2003). Essential role of the cryptic epitope SLAYGLR within osteopontin in a murine model of rheumatoid arthritis. Journal of Clinical Investigation, 112(2), 181-188. doi:10.1172/JCI200317778

Young, M. F. (2003). Bone matrix proteins: Their function, regulation, and relationship to osteoporosis. Osteoporosis International, 14(3), 35-42. doi:10.1007/s00198-002-1342-7

Yuan, F., Xu, M., Li, X., Xinlong, H., Fang, W., & Dong, J. (2016). The roles of acidosis in osteoclast biology. Frontiers in Physiology, 7, 222. doi:10.3389/fphys.2016.00222

Yumoto, K., Ishijima, M., Rittling, S. R., Tsuji, K., Tsuchiya, Y., Kon, S., . . . Noda, M. (2002a). Osteopontin deficiency protects joints against destruction in anti-type II collagen antibody-induced arthritis in mice. Proceedings of the National Academy of Sciences, 99(7), 4556-4561. doi:10.1073/pnas.052523599

Yumoto, K., Ishijima, M., Rittling, S. R., Tsuji, K., Tsuchiya, Y., Kon, S., . . . Noda, M. (2002b). Osteopontin deficiency protects joints against destruction in anti-type II collagen antibody-induced arthritis in mice. Proceedings of the National Academy of Sciences USA, 99(7), 4556-4561. doi:10.1073/pnas.052523599

Zhang, F. J., Gao, S. G., Cheng, L., Tian, J., Xu, W. S., Luo, W., . . . Lei, G. H. (2013). The effect of hyaluronic acid on osteopontin and CD44 mRNA of fibroblast-like synoviocytes in patients with osteoarthritis of the knee. Rheumatology International, 33(1), 79-83. doi:10.1007/s00296-011-2339-3

Zhang, F., Luo, W., Li, Y., Gao, S., & Lei, G. (2015). Role of osteopontin in rheumatoid arthritis. Rheumatology International, 35(4), 589-595. doi:10.1007/s00296-014-3122-z

Zhao, L., Huang, L., & Zhang, X. (2016). Osteoimmunology: Memorandum for rheumatologists. Science China Life Sciences, 59(12), 1241-1258. doi:10.1007/s11427-016-5105-7

Page 102: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

100

Page 103: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

101

Original publications

I. Luukkonen, J., Pascual, L. M., Patlaka, C., Lång, P., Turunen, S., Halleen, J., Nousiainen, T., Valkealahti, M., Tuukkanen, J., Andersson, G., Lehenkari, P. (2017). Increased amount of phosphorylated proinflammatory osteopontin in rheumatoid arthritis synovia is associated to decreased tartrate-resistant acid phosphatase 5B/5A ratio. Plos One, 12(8). doi:10.1371/journal.pone.0182904.

II. Luukkonen, J., Hilli, M., Nakamura, M., Ritamo, I., Valmu, L., Kauppinen, K., Tuukkanen J., Lehenkari, P. (2019). Osteoclasts secrete osteopontin into resorption lacunae during bone resorption. Histochemistry and Cell Biology. doi:10.1007/s00418-019-01770-y.

III. Luukkonen J., Huhtakangas J., Turunen S., Tuukkanen J., Vuolteenaho O., Lehenkari P. (2019). Bone resorption and osteoclastogenesis are stimulated by synovial fluid from osteoarthritis and rheumatoid arthritis patients in vitro. Manuscript.

I and II are published under Creative Commons Attribution 4.0 International

License.

Original publications are not included in the electronic version of the dissertation.

Page 104: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

102

Page 105: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

A C T A U N I V E R S I T A T I S O U L U E N S I S

Book orders:Granum: Virtual book storehttp://granum.uta.fi/granum/

S E R I E S D M E D I C A

1516. Väyrynen, Otto (2019) Factors affecting aggressive oral tongue cancer invasionand development of in vitro models for chemoradiotherapy assay

1517. Euro, Ulla (2019) Risk factors for sciatica

1518. Lotvonen, Sinikka (2019) Palvelutaloon muuttaneiden ikääntyneiden fyysinentoimintakyky, sen muutos ja toimintakykyyn yhteydessä olevat tekijät ensim-mäisen asumisvuoden aikana

1519. Tuomikoski, Anna-Maria (2019) Sairaanhoitajien opiskelijaohjausosaaminen jaohjaajakoulutuksen vaikutus osaamiseen

1520. Raza, Ghulam Shere (2019) The role of dietary fibers in metabolic diseases

1521. Tiinanen, Suvi (2019) Methods for assessment of autonomic nervous systemactivity from cardiorespiratory signals

1522. Skarp, Sini (2019) Whole exome sequencing in identifying genetic factors inmusculoskeletal diseases

1523. Mällinen, Jari (2019) Studies on acute appendicitis with a special reference toappendicoliths and periappendicular abscesses

1524. Paavola, Timo (2019) Associations of low HDL cholesterol level and prematurecoronary heart disease with functionality and phospholipid composition of HDLand with plasma oxLDL antibody levels

1525. Karki, Saujanya (2019) Oral health status, oral health-related quality of life andassociated factors among Nepalese schoolchildren

1526. Szabo, Zoltan (2019) Modulation of connective tissue growth factor and activinreceptor 2b function in cardiac hypertrophy and fibrosis

1527. Karttunen, Markus (2019) Lääkehoidon turvallinen toteuttaminen ikääntyneidenpitkäaikaishoidossa hoitohenkilöstön arvioimana

1528. Honkanen, Tiia (2019) More efficient use of HER targeting agents in cancertherapy

1529. Arima, Reetta (2019) Metformin, statins and the risk and prognosis ofendometrial cancer in women with type 2 diabetes

1530. Sirniö, Kai (2019) Distal radius fractures : Epidemiology, seasonal variation andresults of palmar plate fixation

1531. Näpänkangas, Juha (2019) Pathogenesis of calcific aortic valve disease

Page 106: OULU 2019 D 1532 UNIVERSITY OF OULU P.O. Box 8000 FI-90014

UNIVERSITY OF OULU P .O. Box 8000 F I -90014 UNIVERSITY OF OULU FINLAND

A C T A U N I V E R S I T A T I S O U L U E N S I S

University Lecturer Tuomo Glumoff

University Lecturer Santeri Palviainen

Senior research fellow Jari Juuti

Professor Olli Vuolteenaho

University Lecturer Veli-Matti Ulvinen

Planning Director Pertti Tikkanen

Professor Jari Juga

University Lecturer Anu Soikkeli

Professor Olli Vuolteenaho

Publications Editor Kirsti Nurkkala

ISBN 978-952-62-2363-6 (Paperback)ISBN 978-952-62-2364-3 (PDF)ISSN 0355-3221 (Print)ISSN 1796-2234 (Online)

U N I V E R S I TAT I S O U L U E N S I S

MEDICA

ACTAD

D 1532

AC

TAJani Luukkonen

OULU 2019

D 1532

Jani Luukkonen

OSTEOPONTIN AND OSTEOCLASTSIN RHEUMATOID ARTHRITIS AND OSTEOARTHRITIS

UNIVERSITY OF OULU GRADUATE SCHOOL;UNIVERSITY OF OULU,FACULTY OF MEDICINE;MEDICAL RESEARCH CENTER OULU