Biology of Osteoarthritis

advertisement
ACVP 2014
Biology of Osteoarthritis
Richard F. Loeser, Jr., MD
The Herman and Louise Smith Distinguished Professor
Division of Rheumatology, Allergy, and Immunology
Director of Basic and Translational Research
The University of North Carolina School of Medicine
Chapel Hill, NC 27599
richard_loeser@med.unc.edu
Osteoarthritis is by far the most common form of arthritis affecting both man and the majority of animal
species that have been studied. Currently, there are no known treatments proven to slow or stop
progression of the disease. Therefore, research in the field has focused on elucidating the basic
mechanisms of the disease with the hope that new therapeutic targets will be discovered. Our
understanding of osteoarthritis (OA) has progressed from the old view that it is a non-inflammatory
form of degenerative joint disease resulting from excessive wear and tear on the joints, to the current
concept of excessive remodeling of joint tissues in response to abnormal joint mechanics, systemic
factors, and genetics and driven by localized inflammatory mediators 1-3. Rather than just due to
cartilage breakdown and loss, it is clear that OA is a condition affecting the joint as an organ4 (Fig.1).
The destruction of the articular cartilage is a key
feature and certainly contributes to a loss in normal
joint function but, since cartilage is aneural, cartilage
destruction does not directly result in pain. Other
joint structures affected in OA include the synovium,
bone, menisci (knee), ligaments, joint capsule, bursa,
and local muscle and tendons. These structures are
all potential sources of pain and are probably
affected to a variable extent in each individual
person (or animal). Recent MRI studies in humans
have suggested that synovitis and bone marrow
lesions may be key sources of pain5. The variability in
how the joint tissues are affected is likely the result
of the multifactorial nature of OA.
Synovitis is often present in people with OA but the severity ranges from mild to severe. In a study of
patients with OA severe enough to require joint replacement6, one third had little or no synovitis, one
third moderate synovitis, and one third severe synovitis, suggesting there may be subgroups of people
with OA where synovial inflammation plays a key role. The synovium can be a source of inflammatory
mediators that contributes to the destruction of other joint tissues and progression of disease7 as well as
an important source of pain. A host of inflammatory mediators have been identified in the synovial fluid
of humans with OA including members of the complement family and several cytokines and chemokines
including IL-6, VEGF, MCP-1, IP-10, and MIG 8. The finding of multiple members of the complement
family in human synovial fluid led investigators to study the role of complement in mouse models of OA
where they found evidence that the complement pathway is activated in the OA process and promotes
disease progression9. Future therapies may target the inflammatory mediators produced by the
1
synovium in the subsets of people (or animals) where synovitis is more severe but at the present time a
master regulator of inflammation (such as the apparent role of TNFα in rheumatoid arthritis) has not
been identified in OA.
The bone is commonly affected in OA and involvement ranges from the common findings of osteophytes
and subchondral sclerosis to bone marrow lesions detected on MRI that represent areas of active
localized bone remodeling. Pathologically these lesions show evidence of focal necrosis and fibrosis;
however, as detected by MRI they can be reversible. The location of the bone marrow lesions is
associated with areas of increased biomechanical stress such that varus malalignment that results in
increased medial joint loads is associated with medial lesions while valgus malalignment that results in
increased lateral joint loads is associated with lateral lesions. In humans, these lesions are associated
with pain and disease progression10, 11. There is evidence from animal model studies that the changes in
bone may drive the destruction of the overlying cartilage although this does not hold true in all model
systems with some showing a disconnect between changes in bone and cartilage3, 4. There may be a
subset of people (or animals) with OA where the bone changes are driving disease progression making
bone a potential therapeutic target. But if the bone lesions are only the result of abnormal mechanics
and by themselves do not affect disease progression, then targeting bone without changing the
mechanics will likely be futile.
In the human knee, MRI research studies have found that the menisci are often affected, even in people
without a history of known knee injury, with some type of meniscal change almost always present in
symptomatic knee OA 12-14. Meniscal injury can be a risk factor for the development of OA and as OA
develops from other causes, the meniscus, like the cartilage, appears to be a very vulnerable tissue.
Damage to the meniscus can not only promote OA through changes in joint mechanics with increased
contact stress in areas of meniscus loss but also through the release of inflammatory mediators that are
produced by meniscal cells15, 16.There is no evidence that current surgical techniques used to repair a
damaged or degenerative meniscus are of any benefit in slowing the development of OA. Future
interventions will likely include meniscal replacements using tissue engineering approaches. Likewise,
damage to ligaments, notably the ACL in the knee, is very common in older adults with knee OA. Similar
to the findings of meniscal lesions, many of those with torn or severely degenerated ACLs did not have a
history of trauma17. It appears that degenerative changes in the menisci and ligaments occurs commonly
during the development of OA and their loss of function in turn contribute to the further progression of
OA by altering normal joint mechanics.
The cartilage is still the tissue in the joint that receives the most attention in terms of basic research
studies and is still considered to be an important target for therapy. Cartilage destruction is a key
feature of OA. Often thought to be the shock absorber of the joint (which it is not, it merely distributes
load) articular cartilage is mostly responsible for the normal smooth gliding motion of the joint.
Hyaluronic acid is not the substance responsible for reducing friction on the joint surface but rather a
large mucinous protein called lubricin (also known as superficial zone protein or PRG4), made by the
superficial zone chondrocytes and the synovium, appears to be much more important18. Recombinant
lubricin that can be injected into the joint has been developed and tested in rats19. Trials in humans are
likely to be seen soon and hopefully will show more benefit that HA injections.
The basic mechanisms mediating cartilage destruction in OA can be activated by abnormal forces acting
on normal cartilage or normal forces acting on abnormal cartilage. Abnormal cartilage can result from
either genetic mutations that affect a specific cartilage matrix protein (such as collagen mutations),
metabolic abnormalities (such as hemochromatosis or ochrinosis) or aging changes in the matrix (such
as the accumulation of advanced glycation end-products, matrix fragments and mineral). It is well
2
accepted that the chondrocyte (the one cell type in cartilage) is responsible for the destruction of its
own matrix through the release of matrix degrading enzymes that include several matrix
metalloproteinases, aggrecanases, and cysteine and serine proteases. Evidence is accumulating that the
production of these enzymes is driven by a host of inflammatory mediators including multiple cytokines
(IL-1, IL-6, IL-7, IL-8, IL-17, TNFα, and so on), chemokines (MCP-1, GRO, LIF and others), several
prostaglandins and leukotrienes, as well as reactive oxygen species 4, 20, 21. Aging contributes to the
development of OA in part due to the presence of increased levels of reactive oxygen species resulting
from age-related oxidative stress and due to a decline in autophagy 22-24. Aging chondrocytes become
less responsive to growth factors but maintain their response to cytokines and cartilage matrix
fragments contributing to the imbalance in anabolic and catabolic activity in cartilage and resulting in
cartilage matrix destruction and loss.
Animal models, both spontaneous and injury induced, have been widely used to study OA. Each model
has its advantages and disadvantages and a discussion of the various models is beyond the scope of this
presentation. In order to be able to take advantage of the numerous transgenic and knock-out animals,
much recent attention has been placed on using mouse models with the most commonly accepted
model currently being the destabilized medial meniscus (DMM) model first described by Sonya
Glasson25, 26. In this model, the medial meniscotibial ligament is transected resulting in biomechanical
changes in the joint that lead to progressive pathology that is similar to the pathology seen in human
OA. We have characterized the pathology in this model and examined changes in gene expression using
RNA extracted from joint tissue and microarrays27-30. An important finding from these studies has been
that the age of the animals at the time OA is induced surgically influences both the severity of the
disease and the pattern of gene expression. Mice that were 12 months-old at the time of surgery had
almost twice as severe lesions 8 weeks later when compared to mice that were 12 weeks-old when
DMM surgery was performed28. Importantly, there were striking differences in gene expression changes
with only 55 genes showing a similar expression pattern in young and older adult mice while 493 genes
showed differential expression with many more up-regulated genes in the older mice. Most
investigators using the DMM model use mice between 6-10 weeks of age. Given the differences noted in
our study, and the fact mice that are 6-10 weeks old are equivalent to very young teenaged humans
where OA is extremely rare, investigators need to reconsider the age of animals being used to try and
model human OA.
References
1.
2.
3.
4.
5.
6.
3
Goldring MB, Marcu KB. Cartilage homeostasis in health and rheumatic diseases. Arthritis Res
Ther 2009; 11: 224.
Abramson SB, Attur M. Developments in the scientific understanding of osteoarthritis. Arthritis
Res Ther 2009; 11: 227.
Loeser RF. Osteoarthritis year in review 2013: biology. Osteoarthritis Cartilage 2013; 21: 14361442.
Loeser RF, Goldring SR, Scanzello CR, Goldring MB. Osteoarthritis: A disease of the joint as an
organ. Arthritis Rheum 2012; 64: 1697-1707.
Yusuf E, Kortekaas MC, Watt I, Huizinga TW, Kloppenburg M. Do knee abnormalities visualised
on MRI explain knee pain in knee osteoarthritis? A systematic review. Ann Rheum Dis 2010.
Haywood L, McWilliams DF, Pearson CI, Gill SE, Ganesan A, Wilson D, Walsh DA. Inflammation
and angiogenesis in osteoarthritis. Arthritis Rheum 2003; 48: 2173-2177.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
4
Ayral X, Pickering EH, Woodworth TG, Mackillop N, Dougados M. Synovitis: a potential predictive
factor of structural progression of medial tibiofemoral knee osteoarthritis -- results of a 1 year
longitudinal arthroscopic study in 422 patients. Osteoarthritis Cartilage 2005; 13: 361-367.
Sohn DH, Sokolove J, Sharpe O, Erhart JC, Chandra PE, Lahey LJ, Lindstrom TM, Hwang I, Boyer
KA, Andriacchi TP, Robinson WH. Plasma proteins present in osteoarthritic synovial fluid can
stimulate cytokine production via Toll-like receptor 4. Arthritis Res Ther 2012; 14: R7.
Wang Q, Rozelle AL, Lepus CM, Scanzello CR, Song JJ, Larsen DM, Crish JF, Bebek G, Ritter SY,
Lindstrom TM, Hwang I, Wong HH, Punzi L, Encarnacion A, Shamloo M, Goodman SB, WyssCoray T, Goldring SR, Banda NK, Thurman JM, Gobezie R, Crow MK, Holers VM, Lee DM,
Robinson WH. Identification of a central role for complement in osteoarthritis. Nat Med 2011;
17: 1674-1679.
Felson DT, McLaughlin S, Goggins J, LaValley MP, Gale ME, Totterman S, Li W, Hill C, Gale D.
Bone marrow edema and its relation to progression of knee osteoarthritis. Ann Intern Med
2003; 139: 330-336.
Hunter DJ, Zhang Y, Niu J, Goggins J, Amin S, LaValley MP, Guermazi A, Genant H, Gale D, Felson
DT. Increase in bone marrow lesions associated with cartilage loss: a longitudinal magnetic
resonance imaging study of knee osteoarthritis. Arthritis Rheum 2006; 54: 1529-1535.
Hunter DJ, Zhang YQ, Niu JB, Tu X, Amin S, Clancy M, Guermazi A, Grigorian M, Gale D, Felson
DT. The association of meniscal pathologic changes with cartilage loss in symptomatic knee
osteoarthritis. Arthritis Rheum 2006; 54: 795-801.
Englund M, Guermazi A, Gale D, Hunter DJ, Aliabadi P, Clancy M, Felson DT. Incidental meniscal
findings on knee MRI in middle-aged and elderly persons. N Engl J Med 2008; 359: 1108-1115.
Englund M, Guermazi A, Roemer FW, Aliabadi P, Yang M, Lewis CE, Torner J, Nevitt MC, Sack B,
Felson DT. Meniscal tear in knees without surgery and the development of radiographic
osteoarthritis among middle-aged and elderly persons: The Multicenter Osteoarthritis Study.
Arthritis Rheum 2009; 60: 831-839.
Brophy RH, Rai MF, Zhang Z, Torgomyan A, Sandell LJ. Molecular analysis of age and sex-related
gene expression in meniscal tears with and without a concomitant anterior cruciate ligament
tear. J Bone Joint Surg Am 2012; 94: 385-393.
Stone AV, Loeser RF, Vanderman KS, Long DL, Clark SC, Ferguson CM. Pro-inflammatory
stimulation of meniscus cells increases production of matrix metalloproteinases and additional
catabolic factors involved in osteoarthritis pathogenesis. Osteoarthritis Cartilage 2013.
Hasegawa A, Otsuki S, Pauli C, Miyaki S, Patil S, Steklov N, Kinoshita M, Koziol J, D'Lima DD, Lotz
MK. Anterior cruciate ligament changes in the human knee joint in aging and osteoarthritis.
Arthritis Rheum 2012; 64: 696-704.
Jay GD, Torres JR, Warman ML, Laderer MC, Breuer KS. The role of lubricin in the mechanical
behavior of synovial fluid. Proc Natl Acad Sci U S A 2007; 104: 6194-6199.
Flannery CR, Zollner R, Corcoran C, Jones AR, Root A, Rivera-Bermudez MA, Blanchet T, Gleghorn
JP, Bonassar LJ, Bendele AM, Morris EA, Glasson SS. Prevention of cartilage degeneration in a rat
model of osteoarthritis by intraarticular treatment with recombinant lubricin. Arthritis Rheum
2009; 60: 840-847.
Goldring MB. Osteoarthritis and cartilage: the role of cytokines. Curr Rheumatol Rep 2000; 2:
459-465.
Loeser RF. Molecular mechanisms of cartilage destruction: Mechanics, inflammatory mediators,
and aging collide. Arthritis Rheum 2006; 54: 1357-1360.
Loeser RF. Aging and osteoarthritis: the role of chondrocyte senescence and aging changes in
the cartilage matrix. Osteoarthritis Cartilage 2009; 17: 971-979.
Lotz M, Loeser RF. Effects of aging on articular cartilage homeostasis. Bone 2012; 51: 241-248.
24.
25.
26.
27.
28.
29.
30.
5
Loeser RF. Aging processes and the development of osteoarthritis. Curr Opin Rheumatol 2013;
25: 108-113.
Glasson SS, Askew R, Sheppard B, Carito BA, Blanchet T, Ma HL, Flannery CR, Kanki K, Wang E,
Peluso D, Yang Z, Majumdar MK, Morris EA. Characterization of and osteoarthritis susceptibility
in ADAMTS-4-knockout mice. Arthritis Rheum 2004; 50: 2547-2558.
Glasson SS, Blanchet TJ, Morris EA. The surgical destabilization of the medial meniscus (DMM)
model of osteoarthritis in the 129/SvEv mouse. Osteoarthritis Cartilage 2007; 15: 1061-1069.
McNulty MA, Loeser RF, Davey C, Callahan MF, Ferguson CM, Carlson CS. Histopathology of
naturally occurring and surgically induced osteoarthritis in mice. Osteoarthritis Cartilage 2012;
20: 949-956.
Loeser RF, Olex A, McNulty MA, Carlson CS, Callahan M, Ferguson C, Chou J, Leng X, Fetrow JS.
Microarray analysis reveals age-related differences in gene expression during the development
of osteoarthritis in mice. Arthritis Rheum 2012; 64: 705-717.
Loeser RF, Olex AL, McNulty MA, Carlson CS, Callahan M, Ferguson C, Fetrow JS. Disease
progression and phasic changes in gene expression in a mouse model of osteoarthritis. PLoS One
2013; 8: e54633.
Olex AL, Turkett WH, Fetrow JS, Loeser RF. Integration of gene expression data with networkbased analysis to identify signaling and metabolic pathways regulated during the development
of osteoarthritis. Gene 2014; 542: 38-45.
Download