Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Animal models of rheumatoid arthritis

Richard Williams • Louise M. Topping 95


Key Points arachidonic acid metabolites play an important pathological role in the dis-
n Arthritis may be induced in animals by immunization with cartilage components, ease process. Thus, cyclooxygenase inhibitors are very effective in reducing
nonspecific immune stimuli, localization of antigens in the joint, or genetic mutations. the severity of CIA but are less effective in RA.
n Animal models are tools that mimic various aspects of rheumatoid arthritis (RA) but
As discussed above, convincing data have not yet emerged pointing
do not fully resemble human disease.
definitively to a role for collagen autoimmunity in the bulk of patients with
RA. This has prompted the search for other potential joint antigens that
n Models have a defined onset and are useful for studying initiating factors and kinetic
may be the target of the autoimmune response in RA. For example, a T cell–
evaluation of disease processes.
driven arthritis has been described in BALB/c mice following immunization
n Animal models serve an important role in the evaluation of antiarthritic drugs but do
with the G1 domain of human proteoglycan aggrecan.15,16 Histopathological
not always predict efficacy in man.
changes include edema, proliferative synovitis, infiltration of mononuclear
n There is increasing emphasis on implementation of the 3 R’s: replacement, reduction, cells, pannus formation, and erosion of cartilage and bone.
and refinement.

RESPONSE TO NONSPECIFIC IMMUNOLOGIC STIMULI


INTRODUCTION
Injection of nonspecific agents with adjuvant activity (the capacity to elicit
Experimental animal models of arthritis have contributed to the understand- an indirect immunologic response) can provoke experimental arthritis in
ing of basic mechanisms of joint disease. Marked diversity is seen among the certain species. The classic example is adjuvant arthritis (AA) in the Lewis
numerous models, with arthritis being induced by the following: rat.17 The precise contributions of the oil and the mycobacterial components
n Immunization with cartilage components of Freund’s complete adjuvant in the pathologic pathway of this experimen-
n Injection of nonspecific immune stimuli tal disease remain unclear, but both may contribute. An association between
n Components of infectious agents immunity to 65-kDa heat shock proteins and the induction of AA has been
n Immune complexes suspected,18 but no single mycobacterial immunogen has been shown to be
n Manipulation of genetic information in transgenic animals responsible.19 There is, however, convincing evidence of a role for a myco-
n Spontaneous mutations bacterial cell wall component, muramyl dipeptide, which is immunostimu-
No single animal model of arthritis truly represents the human disease. latory but does not evoke a specific immune response.20
The wide variety of agents that can induce experimental arthritis with clinical
and histopathologic features close to those of human arthritides suggests that Adjuvant- and pristane-induced arthritis
disparate etiologic pathways could exist for rheumatoid arthritis (RA). Aspects In addition, a number of adjuvants that lack immunogenic properties have
peculiar to individual models are of value but must be interpreted with cau- been shown to induce arthritis in susceptible strains of rats, including avri-
tion. Much can be learned from the general validity of mediator involvement dine,21 incomplete Freund’s adjuvant, and pristane.19 Arthritis has also been
and common concepts. Models provide valuable preclinical data for the devel- observed in mice following administration of pristane.22 Confirmation of
opment of novel treatments, both pharmacologic and biologic, and insight the fact that these are T cell–driven diseases is provided by the fact that
into relevant mechanisms common to both experimental arthritis and RA. (1) anti–T cell treatments prevent the induction of arthritis,19,23 (2) suscep-
tibility is associated with genes within the MHC,24,25 and (3) arthritis may
be adoptively transferred by T cells.26,27 It was also found that arthritis could
MECHANISMS OF INDUCTION OF be induced in DA rats by percutaneous exposure of adjuvant oils28 or even
a mineral oil–containing cosmetic product.29 The mechanism of arthritis
EXPERIMENTAL ARTHRITIS induction following immunization with adjuvants is not fully understood,
but one possibility is that immunization leads to an increase in the activity
IMMUNIZATION WITH CARTILAGE COMPONENTS of APC,30 resulting in the presentation to T cells of a hitherto unrecognized
The most widely utilized mode of RA is collagen-induced arthritis (CIA), or “sequestered” endogenous antigen.
which occurs in rats, mice, and primates following immunization with type Expansion of autoreactive T- and B-cell populations can occur as a con-
II collagen in adjuvant. The pathological features of the disease include sequence of the adjuvant activation process and result in the migration of
synovitis with infiltration of polymorphonuclear and mononuclear cells, cells to the joint, leading to subsequent immune-mediated joint damage and
pannus formation, erosion of bone and cartilage, and fibrosis (Fig. 95.1). production of a spectrum of autoantibodies against cartilage antigens (Fig.
CIA susceptibility is linked to the I-A region of the H-2q and H-2r hap- 95.2). Disease onset is rapid in AA (2 weeks), often followed by spontaneous
lotypes in mice. Analysis of the I-A chains of alleles from susceptible and remission. Pristane-induced arthritis is slower (months) and characterized by
resistant strain (B10.Q) indicated that susceptibility is associated with a remissions and relapses, with no indication of B-cell involvement at the onset.
four–amino-acid sequence in the I-Aβ chain.1 This sequence is located in a The pristane model is highly suited to studying genes controlling onset, sever-
region associated with binding antigenic peptide, analogous to the genetic ity, and chronicity.31 The spontaneous remission and lack of susceptibility to
susceptibility observed in RA in humans conferred by the DRβ chain. The reinduction make AA a suitable model for studies on the regulation of T-cell
induction of arthritis in mice of a C57BL/6 (H-2b) background2,3 has facil- tolerance. Histopathologic analysis of AA shows major involvement of bone
itated the use of gene knockout mice and the generation of the congenic marrow, bone erosion, extensive bone apposition, and minor direct cartilage
C57Bl/6 N.Q strain that expresses the arthritis susceptible Q haplotype of damage in the early stages (Table 95.1). Indirect cartilage damage occurs later,
the MHC class II region.4 mainly as a consequence of the loss of underlying bone. The latter may explain
Humoral immune responses play a significant role in the pathogenesis the cartilage-protective effect of treatment with osteoprotegerin (OPG), which
of CIA,5 which is likely to be true also for human RA, although elevated is a selective inhibitor of the osteoclast activator RANKL (receptor activator of
circulating levels of antibodies to type II collagen are not detected in the nuclear factor κB ligand).32,33 Combined blocking of TNF and IL-1 is associ-
majority of patients with RA. Another important similarity between CIA ated with optimal control of arthritis and joint destruction.
and RA is the expression of proinflammatory cytokines, including tumor
necrosis factor-alpha (TNF-α) and interleukin (IL)-1β, in the joints of mice
with arthritis6 and the fact that blockade of these molecules results in reduc-
COMPONENTS OF INFECTIOUS AGENTS
tions in both the clinical and histological severity of disease.7–14 However, The development of arthritis as a consequence of infection is apparent in
CIA differs from human RA in that it is a relatively acute disease in which both natural and induced animal models. Infectious agents may be tropic for

799
Descargado para Grecia Vera Vallejos (greciav.9@hotmail.com) en Ricardo Palma University de ClinicalKey.es por Elsevier en junio 15, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
800 SECTION 7  Rheumatoid Arthritis

HYPOTHETICAL MECHANISM FOR THE DEVELOPMENT OF COLLAGEN-INDUCED AND PROTEOGLYCAN-INDUCED EXPERIMENTAL ARTHRITIS

High-density Immune-mediated
Cartilage aggrecan Type II joint damage
Autoantibodies
components proteoglycans collagen Antigen T-cell recognition
Immunization presentation (restricted by
(adjuvant) (MHC restricted) T-cell repertoire)
Th2
Chronic
arthritis
Th1,
Th17

Autoreactive T cells

FIG. 95.1  Animals are immunized with type II collagen or high-density aggrecan proteoglycans, and an autoimmune response results when specific epitopes are presented in
the context of major histocompatibility complex (MHC) class II antigens and recognized by autoreactive CD4+ T-cell populations. Autoantibodies and T effector cells reactive
against cartilage components develop via helper T-cell pathways and localize in synovial joints, which causes an immune-mediated inflammatory response. The immune
response is perpetuated by the release of cartilage antigens within the damaged joint, and chronic arthritis develops.

be induced in Lewis rats34 by the systemic injection of cell wall fragments


HYPOTHETICAL MECHANISM FOR THE DEVELOPMENT OF PRISTANE- of group A streptococci, which are highly resistant to biodegradation. A
INDUCED AND ADJUVANT-INDUCED EXPERIMENTAL ARTHRITIS similar disease can be induced with fragments from other bacteria, such as
Lactobacillus casei or Eubacterium aerofaciens. The underlying principle of
Chronic antigen this model resides in the poor degradability of the fragments, thereby creat-
stimulation ing a persistent stimulus.
The Lactobacillus and Eubacterium models are of particular interest for
Joint antigen human disease because these bacteria are part of the normal gastrointestinal
Autoreactive and heat shock flora. An enormous amount of potential arthritogenic stimuli is continu-
B-cell activation protein release ously present in the normal gastrointestinal tract; fragments may spread in a
noninfectious manner to other tissues, and this may provoke arthritis when
immunoregulatory control is insufficient.
Within 24 hours of administration of cell wall fragments to rats, acute
Macrophage Immune- inflammation develops in peripheral joints, coincident with dissemina-
stimulation mediated
(pristane, adjuvants)
tion of the cell wall fragments in blood vessels of the synovium and sub-
joint damage chondral bone marrow. The acute, complement-dependent inflammation
subsides over the next week, followed within 2 weeks by a chronic, T cell–
Autoreactive dependent erosive polyarthritis involving mainly peripheral joints. Chronic
Cytokine T-cell activation joint inflammation develops only in susceptible strains that are unable to
production (MHC and Mls restricted)
maintain tolerance and therefore display SCW-specific T-cell responses.
Lewis rats are highly vulnerable to this arthritis. The mouse strains studied
thus far are not susceptible to the single intraperitoneal injection model, and
repeated dosing is needed to break tolerance. It is tempting to speculate that
Chondrocyte activation
similar loss of tolerance may occur in patients with RA with age, combined
with costimulatory environmental factors.
Studies of the involvement of cytokines in SCW arthritis show a combined
Synovial cell activation role of TNF and IL-1, as found in AA.35 In mice, a chronic relapsing SCW
model can be induced by repeated weekly injection of SCW fragments directly
FIG. 95.2  Circulating macrophages become intensely activated by the nondegrad- into the knee joint, which displays a gradually increasing role of T cell–derived
able oil components and produce high levels of proinflammatory cytokines (inter- IL-17 and synovial IL-17 receptor–bearing cells with every flare.36
leukin-1 and interleukin-6, tumor necrosis factor-α, and interferon-α). Genetic Bacterial DNA can induce arthritis. In particular, the CpG motifs are
regulation governs the presence of autoreactive T cells and B cells, which become arthritogenic, and substantial amounts can be found in joint tissues.37
activated and clonally expand. Joint-reactive lymphocytes migrate to the joint and Macrophages play a major role in this arthritis through TNF production.
induce immune-mediated joint damage. The release of cartilage antigens and heat However, in comparison to cell wall fragments, the cytokine-inducing
shock proteins may feed back to the autoreactive lymphocytes and result in chronic capacity of bacterial DNA is weak. Normal joints of individuals contain both
immune stimulation. Cytokines may also act directly on both synovial cells and bacterial fragments and bacterial CpG (CP61) motifs, and it is conceivable
chondrocytes and thereby result in synovial hypertrophy, abnormal expression of that both factors contribute to arthritis.
major histocompatibility complex (MHC) class II antigens and adhesin molecules,
and atypical production of matrix components. Arthritis develops as a result of the Antigen-induced arthritis
chronic nature of the inflammatory stimulation. Antigen-induced arthritis (AIA) provides a designed model of the develop-
ment of chronic erosive arthritis that reflects a sustained immune reaction to
a persistent trigger in joint tissues, in this case a planted protein antigen. It is
the joint as a result of expression of adhesins or other molecules that pro- elicited by local injection of a high dose of antigen into the knee joint of an
mote sequestration within synovial tissue. Bacterial cell wall and Mycoplasma animal previously hyperimmunized with that antigen in Freund’s complete
membrane components may also provide nonspecific immune activation adjuvant. Such a model was first developed by Dumonde and Glynn38 in
through mitogenic activity. Viral infection of synovial cells may elicit novel rabbits. In principle, it can be induced in any species, provided that proper
cell-surface antigens or abnormal expression of activation antigens and the immunity to a particular antigen can be mounted. Applications have since
overproduction of autoantigens. Viral infection may also disrupt immune been developed in mice, rats, and guinea pigs.
regulation and increase proinflammatory cytokines and immune activity In contrast to the polyarthritis models described thus far, AIA remains
against normally immunologically privileged components of the joint. confined to the injected joint. Commonly used antigens are ovalbumin,
bovine serum albumin (BSA), fibrin, and cationic forms such as methylated
Streptococcal cell wall arthritis BSA. Cationic antigens are preferred due to their increased retention in the
Persistence of antigens from microorganisms within the joint can be critical joint.39 Preimmunization with antigen in complete Freund adjuvant induces
for the induction of arthritis. Streptococcal cell wall (SCW) arthritis can strong humoral and cell-mediated immunity. Arthritis is usually induced 3

Descargado para Grecia Vera Vallejos (greciav.9@hotmail.com) en Ricardo Palma University de ClinicalKey.es por Elsevier en junio 15, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
CHAPTER 95  Animal models of rheumatoid arthritis 801

Table 95.1
Models of Arthritis
Model Abbreviation Speciesa Feature IC T Cell References
Induced models
Collagen-induced arthritis CIA DBA/1 mouse CII + + 73

Proteoglycan-induced arthritis PGA BALB/c mouse PG + + 74

Adjuvant-induced arthritis AA Lewis rat Autoimmune − + 17

Oil-induced arthritis OIA DA rat Autoimmune − + 19

Pristane-induced arthritis PIA Mouse, DA rat Autoimmune − + 75,76

Streptococcal cell wall–induced arthritis SCW-A Lewis rat Persistent bacteria − + 34

Flare SCW Mouse T cell–derived IL-17 − + 35

Antigen-induced arthritis AIA Rabbit, mouse Persistent antigen + + 38,40

Flare AIA Mouse T cell–derived IL-17 − + 41

Transgenic models
K/BxN arthritis KRN K/BxN mouse GPI + + 46

TNF transgenic arthritis TNFtg Mouse TNF overexpression − − 61,62

IL-1 transgenic arthritis IL-1tg Mouse IL-1 overexpression − − 65

IL-1ra transgenic arthritis IL-1ra−/− BALB/c mouse Autoimmune T cells ± + 69,71

SKG arthritis SKG Mouse T-cell defect − + 58

gp130-induced arthritis gp130 Mouse STAT3 defect − + 60

gp130, Glycoprotein 130; GPI, glucose-6-phosphate isomerase; IC, immune complexes; IL-1, interleukin 1; TNF, tumor necrosis factor.
a
Mostly used.

weeks later by local injection of a large amount of antigen into the knee sensitized joint from the circulation. Flares of smoldering arthritis can be
joint. Initially, an immune complex (IC) type of reaction dominates, fol- induced with as little as 10 ng of antigen, and T cell–derived IL-17 plays a
lowed by T cell–mediated chronic inflammation. In the rabbit, chronicity dominant part in such exacerbations.41
may last for years. Histopathologic examination shows a granulocyte-rich Similar flare models have been developed in rats and mice with bacte-
exudate in the joint space, thickening of the synovial lining layer, and, at rial cell wall constituents. Bacterial fragments may function as an antigen
later stages, a predominantly mononuclear infiltrate in the synovium, which but also directly trigger TLRs, in particular, TLR2. The ensuing reactions
later includes numerous T cells and clusters of plasma cells making anti- are a mixture of T cell– and macrophage/fibroblast-driven processes. In the
bodies to the inciting antigen, thus implying that retained antigen is still a mouse model of repeated SCW rechallenge, the swelling response of every
driving force in chronic arthritis. Intense IC formation is seen in superficial flare remains dependent on TNF. However, the chronic erosive infiltrate in
layers of the articular cartilage, which may contribute to localized cartilage the synovial tissue that occurs after repeated challenges is dependent on
destruction. Early loss of PG, followed by pannus formation and cartilage IL-1. Cartilage erosion is absent in IL-1-deficient mice but does occur in
and bone erosion, is a common finding. TNF-deficient mice. This model is more severe and erosive in DBA/1 mice
Two important principles emerge from this model: first, chronicity than in C57Bl/6 mice, erosion is absent in T/B cell–deficient RAG−/− mice,
occurs only in the presence of sufficient antigen retention in joint tissues, and blocking of IL-17 prevents an erosive phenotype. The repeated-chal-
in combination with proper T cell–mediated delayed hypersensitivity, and, lenge model becomes Th17 dependent, and IL-1 is a major cytokine driving
second, joints contain numerous noncollagenous and avascular collagenous this process of Th17 generation.36
tissue such as cartilage, ligaments, and tendons, which allows prolonged Of note, considerable cross-reactivity occurs between cell walls from
antigen retention by antibody-mediated trapping and charge-mediated bind- different bacterial origins, and flares may result from homologous and het-
ing.39,40 Chronicity is caused by the generation of local antigen-specific T-cell erologous fragments. This may extend to cross-reactive autoantigens from
hyperreactivity, which allows flares in response to tiny amounts of antigen. cartilage, which underlines the fact that arthritis can start in response to a
In rabbits, antibody responses are generally high and allow sufficient particular antigen but may spread to other antigens, including autoantigens.
IC-mediated trapping of antigen in the joint. Because antibody levels are TLRs have been identified as recognition sites for bacteria. TLR2 is
lower in mice, cationic antigens are needed as suitable arthritogens because involved in SCW recognition, whereas TLR4 is triggered by lipopolysaccha-
of their ability to stick to the negatively charged collagenous structures of ride but also by the damaged connective tissue components released in ero-
the joint and to accumulate IC formation at the surface.39,40 Of interest, this sive stages. The acute SWC arthritis model is dependent on TLR2, whereas
principle may extend to cationic bacterial or viral components and appears the more chronic rechallenge model loses TLR2 dependence and becomes
to be of importance in the KRN model of arthritis, in which anti-glucose-6- dependent on TLR4.42 These principles open up a wide range of putative
phosphate isomerase (anti-GPI) antibodies stick to GPI antigen trapped at stimuli involved in exacerbations, which simultaneously complicates the
cartilage surfaces. search for the driving “antigen” in humans. Flares can be blocked efficiently
In AIA in the rabbit and the mouse, elimination of TNF and IL-1 was with a combination of antibodies to TNF, IL-1, and IL-1741 in particular.
poorly effective in suppressing joint inflammation, thus pointing to substan- Evaluation of the efficacy of TLR blockers is warranted.
tial “overkill” by other mediators in this severe onset of arthritis. However,
elimination of IL-1 did yield impressive protection against cartilage destruc-
tion. The AIA model is most suited to studies of the mechanism of cartilage
IMMUNE COMPLEX–INDUCED ARTHRITIS
destruction as induced by a mix of ICs and T-cell reactivity. It is assisted Autoantibodies such as rheumatoid factor and anticitrullinated peptide anti-
by knowledge of the exact time of onset, accessibility of the knee joint (as bodies (ACPAs) are a key feature of RA, and the success of treatment with an
compared with the ankles), and the presence of a contralateral control joint. anti–B cell drug (rituximab) enhanced interest in their pathogenic role. In
Moreover, the model can be used to evaluate the regulation of local T-cell some of the models discussed earlier such as collagen-, PG-, and antigen-in-
hyperreactivity against a retained foreign antigen. duced arthritis, IC formation at joint tissues is a major element (see Table
95.1). Although excessive IC formation can cause destructive arthritis, chro-
Flares of arthritis nicity is limited, unless augmented by T cells.43 Minute amounts of antigen
In contrast to the chronicity of human RA, most animal models have a rela- suffice to stimulate T cells, whereas considerable amounts of ICs are neces-
tively short duration of severe and rapidly destructive inflammation. In this sary to stimulate the release of inflammatory mediators from phagocytes. It
respect, models of repeated flares of arthritis, with slower development of is likely that IC models mimic part of the RA pathology.
lesions, provide a valuable extension. There is interest in the use of passive IC models, along with the avail-
An arthritic joint bearing retained antigen and a chronic antigen-specific ability of a range of transgenic knockouts, to identify crucial pathways of
T-cell infiltrate displays a state of local hyperreactivity. This situation is not inflammation and tissue destruction. The advantage of passive systems is
restricted to retained antigen but also applies to new antigen entering the the lower dependence on genetic background, thereby avoiding excessive

Descargado para Grecia Vera Vallejos (greciav.9@hotmail.com) en Ricardo Palma University de ClinicalKey.es por Elsevier en junio 15, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
802 SECTION 7  Rheumatoid Arthritis
backcrossing to create transgenics in suitable, susceptible mouse strains.
Passive transfer of collagen-induced arthritis can be performed with a criti-
cal mixture of a number of anti-CII monoclonal antibodies, including com-
plement-binding IgG2a. Sets are now commercially available, and accepted
concepts of inflammation pathways include IC-mediated complement acti-
vation and Fcγ receptor triggering on phagocytes.

K/BxN and serum transfer models


An intriguing model of arthritis was described in the offspring of KRN TCR
transgenic mice crossed with NOD mice.44 KRN TCR transgenic mice express
a TCR specific for bovine pancreas ribonuclease presented in the context of
I-Ak.45 However, when KRN mice are crossed with NOD mice (I-Ag7), the
resulting (K/BxN) offspring develop arthritis spontaneously. Further studies
showed that arthritis in K/BxN mice was dependent on I-Ag7 MHC class II
molecules and CD4+ T cells.44,46,47 B lymphocytes were required for arthri-
tis development,44,46 and arthritis could be transferred by injecting naive
mice with serum IgG from arthritic mice in a complement-dependent and a
FcγR-dependent manner.46,48,49 These antibodies recognize endogenous GPI,
which seems to associate preferentially with the cartilage surface.50
However, the KRN model differs from human RA in one important
respect: its lack of dependence on TNF-α.51 Another point that is worth
emphasizing is that GPI is unlikely to be the critical autoantigen in RA as
antibodies to GPI are no more common in RA than in other diseases.52,53
As an adaptation of this model, Kamradt and colleagues54 immunized
mice with GPI and showed the development of arthritis after a few weeks.
This direct immunization model is a mix of GPI-specific T cells and anti-GPI
antibodies; serum from these mice cannot transfer arthritis. The latter makes
the direct immunization model different from classic KRN arthritis.

ANTICITRULLINATED PROTEIN ANTIBODIES


AND ARTHRITIS MODELS
Anticitrullinated protein antibodies (ACPAs) are a serologic hallmark of
human RA, but their role in animal models of arthritis has been difficult to
confirm. In one study, a range of monoclonal antibodies specific for citrulli- b
nated CII were generated and found to induce arthritis on passive transfer.55
A potential association was identified between periodontal infection and FIG. 95.3 Joint damage in hTNF-α-transgenic mice. (a) Erosive changes in
RA via bacteria-dependent induction of a pathogenic autoimmune response the cartilage–bone–pannus region of a proximal interphalangeal joint from an
to citrullinated epitopes. Thus, infection with the periodontal pathogen hTNF-α-transgenic mouse with arthritis. Note the focal erosion of subchondral bone.
Porphyromonas gingivalis exacerbated CIA, and this was dependent on pepti- (b) Normal joint from a nontransgenic littermate. Hematoxylin and eosin.
dylarginine deiminase (PPAD), which converts arginine residues in proteins
to citrulline. Infection with wild-type P. gingivalis caused increased levels
of autoantibodies to type II collagen type II, whereas a PPAD-null mutant the role of the transgenically expressed TNF-α in the induction of arthritis.
strain was ineffective.56 However, in another study, immunization of various Histopathological analysis of the joints of hTNF-α transgenic mice revealed
mouse strains with citrullinated fibrinogen, myelin basic protein, and type that the disease was highly erosive in nature, with subchondral bone, rather
II collagen failed to induce or aggravate arthritis despite eliciting ACPAs.57 than cartilage, being mostly affected (Fig. 95.3).
TNF-α expression in hTNF-α transgenic mice was not confined to the
joint and was found to be overexpressed in various tissues, including lung,
GENE MANIPULATION AND TRANSGENIC MODELS spleen, and the joint. The reason why the joint should be affected while
Transgenic animals and spontaneous mutant mice have provided models of other tissues are spared is not known and, in fact, a second strain of TNF-α
arthritis, often unexpectedly. For example, Sakaguchi et al. showed that a transgenic mice was generated in which the TNF-α transgene lacked an
spontaneous point mutation of the gene encoding an SH2 domain of ZAP- AU rich region. These mice were found to develop not only arthritis but
70 resulted in the development of chronic autoimmune arthritis in mice.58 also inflammatory bowel disease.62 It is interesting to note that TNF-α-
The result of this is that T cells in SKG mice respond weakly to antigenic overproducing mice can be backcrossed to RAG−/− mice without altering the
stimulation, and it is proposed that suboptimal signal transduction via the arthritis phenotype, indicating that the adaptive immune system plays no
TCR as a result of aberrant ZAP-70 leads to a change in the threshold of T role in the development of arthritis in this model.
cells to thymic selection, resulting in a failure to delete self-reactive T cells. This model is of interest in identifying pathways of TNF-induced arthri-
This model challenges another commonly held belief that autoimmune dis- tis and screening the efficacy of various TNF-directed therapies. A signifi-
eases arise as a result of hyperresponsive T cells. Another interesting facet cant finding was that treatment of hTNF-α transgenic mice with anti-IL-1R
of the SKG model of arthritis is the requirement for stimulation through antibody prevented the development of spontaneous arthritis,63 demon-
the innate immune system, which can be provided by housing the mice in strating that IL-1 is an important downstream pathological mediator in this
a microbially rich environment or by administration of fungal β-glucans, model. This is consistent with findings in human RA synovial cell cultures
such as zymosan.59 This demonstrates how genetic factors can interact with in which TNF-α inhibition was found to block IL-1 production,64 indicating
environmental stimuli to generate autoimmune disease. the dependence of IL-1 production on TNF-α.
Mice with a homozygous mutation in the gp130 IL-6 receptor subunit Another transgenic model that confirms the arthritogenic potential of IL-1
show enhanced signal transduction and STAT3 activation, and a lympho- is the IL-1α-transgenic mouse that spontaneously develops severe arthritis at
cyte-mediated RA-like joint disease develops. The increased proliferation of around one month of age.65 Unlike TNF-α-transgenic mice, severe degrada-
CD4+ T cells is due to elevated production of T cell–activating IL-7 by non- tion of cartilage was observed in IL-1α-transgenic mice, thereby confirming
hematopoietic cells.60 previous findings, published over 30 years ago, regarding the important role
Keffer et al. generated a strain of mice overexpressing a human TNF-α of IL-1 in cartilage breakdown.66–68
transgene (including its endogenous promoter region) that had been dys- The opposite approach, elimination of IL-1 control by gene targeting of
regulated by the replacement of the 3′ AU-rich region with the 3′ untrans- the endogenous IL-1 receptor antagonist (IL-1ra), also yielded a model of
lated region of the human β-globin gene. TNF overexpression resulted in arthritis. IL-1ra deficiency in a BALB/ca background resulted in pronounced
chronic polyarthritis with a 100% incidence.61 The disease could be pre- arthritis at the age of 8 weeks.69 Marked synovial and periarticular inflam-
vented by continuous administration of antihuman TNF-α mAb, confirming mation with invasion of granulation tissue and articular erosion was noted.

Descargado para Grecia Vera Vallejos (greciav.9@hotmail.com) en Ricardo Palma University de ClinicalKey.es por Elsevier en junio 15, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
CHAPTER 95  Animal models of rheumatoid arthritis 803

Moreover, elevated levels of antibodies against immunoglobulins, CII, and 3. Inglis JJ, Criado G, Medghalchi M, et al. Collagen-induced arthritis in C57BL/6 mice is
double-stranded DNA were found, suggestive of autoimmune responses. associated with a robust and sustained T-cell response to type II collagen. Arthritis Res Ther.
Overexpression of a range of cytokines, including IL-1β, TNF, and IL-6, was 2007;9:R113.
4. Backlund J, Li C, Jansson E, et al. C57BL/6 mice need MHC class II Aq to develop collagen-induced
observed in the joints before the onset of arthritis.
arthritis dependent on autoreactive T cells. Ann Rheum Dis. 2013;72(7):1225–1232.
In sharp contrast to the TNF transgenic model, the arthritis in IL-1ra−/− mice 5. Holmdahl R, Andersson ME, Goldschmidt TJ, et al. Collagen induced arthritis as an exper-
is strongly dependent on T cells, in line with the remarkable genetic restriction. imental model for rheumatoid arthritis. Immunogenetics, pathogenesis and autoimmunity.
It is consistent with the view that IL-1 is a crucial regulator of T-cell function. APMIS. 1989;97:575–584.
Undisturbed IL-1 action, in the absence of IL-1ra, apparently permits activation 6. Marinova-Mutafchieva L, Williams RO, Mason LJ, Mauri C, Feldmann M, Maini RN.
of IL-17-producing T cells directed against exogenous triggers or endogenous Dynamics of proinflammatory cytokine expression in the joints of mice with collagen-
autoantigens. The spontaneous arthritis in IL-1ra−/− mice does not develop in induced arthritis (CIA). Clin Exp Immunol. 1997;107:507–512.
germ-free conditions and is reduced in TLR4-deficient mice.70 Both TNF and 7. Thorbecke GJ, Shah R, Leu CH, Kuruvilla AP, Hardison AM, Palladino MA. Involvement of
IL-17 deficiency prevent the onset of arthritis.71 endogenous tumor necrosis factor α and transforming growth factor β during induction of
collagen type II arthritis in mice. Proc Natl Acad Sci U S A. 1992;89:7375–7379.
8. Williams RO, Feldmann M, Maini RN. Anti-tumor necrosis factor ameliorates joint disease in
THE 3 R’s murine collagen-induced arthritis. Proc Natl Acad Sci U S A. 1992;89:9784–9788.
9. Piguet PF, Grau GE, Vesin C, Loetscher H, Gentz R, Lesslauer W. Evolution of collagen
Animal models of arthritis will continue to be used to address scientific arthritis in mice is arrested by treatment with anti-tumour necrosis factor (TNF) antibody
questions and to test novel therapeutic approaches due to the unmet need or a recombinant soluble TNF receptor. Immunology. 1992;77:510–514.
in human disease. The implementation of the 3 R’s—replacement, reduction, 10. Geiger T, Towbin H, Cosenti-Vargas A, et al. Neutralization of interleukin-1β activity in
and refinement—is encouraged at all levels; that is, within research establish- vivo with a monoclonal antibody alleviates collagen-induced arthritis in DBA/1 mice and
ments and companies and by regulators, funding bodies, and journals. There prevents the associated acute-phase response. Clin Exp Rheumatol. 1993;11:515–522.
is considerable scope for the refinement of animal models of RA, including 11. Wooley PH, Dutcher J, Widmer MB, Gillis S. Influence of a recombinant human soluble
tumour necrosis factor receptor Fc fusion protein on type II collagen-induced arthritis in
the use of analgesia, use of less severe models (e.g., not requiring the use of
mice. J Immunol. 1993;151:6602–6607.
CFA), and the inclusion of humane endpoints and improved husbandry.72 12. Van den Berg WB, Joosten LA, Helsen M, van de Loo FA. Amelioration of established murine
Refinement in RA research can easily be employed through the choice of collagen-induced arthritis with anti-IL-1 treatment. Clin Exp Immunol. 1994;95:237–243.
model used. Polyarthritis models—that is, models in which multiple joints 13. Williams RO, Ghrayeb J, Feldmann M, Maini RN. Successful therapy of collagen-

are affected, such as CIA and K/BxN—are associated with an overall higher induced arthritis with TNF receptor-IgG fusion protein and combination with anti-CD4.
severity than monoarthritic models, such as AIA.72 The involvement of only Immunology. 1995;84:433–439.
one joint means mice can redistribute their weight to the three unaffected 14. Joosten LAB, Helen MMA, van de Loo FAJ, Van den Berg WB. Anticytokine treatment of
limbs. Furthermore, the presence of an unaffected contralateral joint acts established type II collagen-induced arthritis in DBA/1 mice: a comparative study using
as an internal control in these models, which for many readouts reduces anti-TNFα, anti-IL-1α/β, and IL-1Ra. Arthritis Rheum. 1996;39:797–809.
15. Glant TT, Mikecz K, Arzoumanian A, Poole AR. Proteoglycan-induced arthritis in BALB/c
the need for additional mice as controls. Although polyarthritis models are
mice. Clinical features and histopathology. Arthritis Rheum. 1987;30:201–212.
scientifically justified due to their apparent similarity to human RA, monoar- 16. Glant TT, Fulop C, Mikecz K, Buzas E, Molnar G, Erhardt P. Proteoglycan-specific autore-
thritis models possess many relevant disease processes and are encouraged active antibodies and T-lymphocytes in experimental arthritis and human rheumatoid joint
to be used in place of polyarthritis models wherever possible. diseases. Biochemical Soc Trans. 1990;18:796–799.
The use of analgesia is also highly encouraged to reduce pain and suffer- 17. Pearson CM. Development of arthritis, periarthritis and periostitis in rats given adjuvants.
ing in RA models but is often not employed due to the lack of literature asso- Proc Soc Exp Biol Med. 1956;91:95–101.
ciated with model progression and drug interaction. Analgesics that inhibit 18. van Eden W, Thole JE, Van der Zee R, Noordzij A, van Embden JD, Cohen IR. Cloning
COX-2 enzymes, such as nonsteroidal antiinflammatories, are not appro- of the mycobacterial epitope recognized by T lymphocytes in adjuvant arthritis. Nature.
priate for use due to their interference with inflammation and the model 1988;331:171–173.
19. Holmdahl R, Goldschmidt TJ, Kleinau S, Kvick C, Jonsson R. Arthritis induced in rats with
progression. Recommended analgesics include the nonopioid analgesic met-
adjuvant oil is a genetically restricted, alpha beta T-cell dependent autoimmune disease.
amizole and aniline analgesics, and there is growing encouragement for their Immunology. 1992;76:197–202.
use as rescue therapy in animal models. 20. Kohashi O, Aihara K, Ozawa A, Kotani S, Azuma I. New model of a synthetic adjuvant,
The use of CFA in animal models is associated with a variety of side N-acetylmuramyl-L-alanyl-D-isoglutamine-induced arthritis: clinical and histologic studies
effects, including localized injection site granulomas and ulceration. The in athymic nude and euthymic rats. Lab Invest. 1982;47(1):27–36.
immunostimulatory ability of CFA means it is still commonly used in RA 21. Chang YH, Pearson CM, Abe C. Adjuvant polyarthritis. IV. Induction by a synthetic adju-
murine models; however, researchers are encouraged to explore alternatives vant: immunologic, histopathologic, and other studies. Arthritis Rheum. 1980;23(1):62–71.
to CFA to reduce pain and suffering in mice. If CFA is to be employed, 22. Bedwell AE, Elson CJ, Hinton CE. Immunological involvement in the pathogenesis of
measures can be taken to minimalize ulceration of the injection site, includ- pristane-induced arthritis. Scand J Immunol. 1987;25(4):393–398.
23. Larsson P, Holmdahl R, Dencker L, Klareskog L. In vivo treatment with W3/13 (anti-pan T)
ing subcutaneous injection instead of intradermal, monitoring injection site
but not with OX8 (anti-suppressor/cytotoxic T) monoclonal antibodies impedes the devel-
after immunization, and treating with topical antiseptic creams daily (e.g., opment of adjuvant arthritis in rats. Immunology. 1985;56(3):383–391.
Sudocrem). 24. Vingsbo C, Jonsson R, Holmdahl R. Avridine-induced arthritis in rats; a T cell-dependent
Another important refinement is to limit the duration of arthritis to the chronic disease influenced both by MHC genes and by non-MHC genes. Clin Exp Immunol.
shortest possible time required to answer a given experimental question. A 1995;99:359–363.
broader discussion of potential refinements is included in Hawkins et al.72 25. Lorentzen JC, Klareskog L. Susceptibility of DA rats to arthritis induced with adjuvant oil
or rat collagen is determined by genes both within and outside the major histocompatibility
complex. Scand J Immunol. 1996;44(6):592–598.
CONCLUSIONS 26. Kleinau S, Klareskog L. Oil-induced arthritis in DA rats passive transfer by T cells but not
with serum. J Autoimmun. 1993;6:449–458.
Arthritis may be induced in animals by immunization with cartilage compo-
27. Svelander L, Mussener A, Erlandsson-Harris H, Kleinau S. Polyclonal Th1 cells transfer
nents, nonspecific immune stimuli, bacterial or viral components, or trans- oil-induced arthritis. Immunology. 1997;91(2):260–265.
genic manipulation. Animal models are research tools that mimic various 28. Kleinau S, Erlandsson H, Klareskog L. Percutaneous exposure of adjuvant oil causes arthri-
aspects but never fully resemble human RA. Animal models have a defined tis in DA rats. Clin Exp Immunol. 1994;96:281–284.
onset and are useful for kinetic evaluation of arthritis, cell and mediator 29. Sverdrup B, Klareskog L, Kleinau S. Common commercial cosmetic products induce arthri-
involvement, and detailed analysis of joint erosion. Animal models serve an tis in the DA rat. Env Health Perspect. 1998;106(1):27–32.
important role in the evaluation of antirheumatic treatments and provide 30. Warren HS, Vogel FR, Chedid LA. Current status of immunological adjuvants. Annu Rev
direction for novel approaches to treatments such as cytokine inhibition. Immunol. 1986;4:369–388.
The implementation of the 3 R’s is of particular relevance for animal models 31. Holmdahl R, Lorentzen JC, Lu S, et al. Arthritis induced in rats with nonimmunogenic
adjuvants as models for rheumatoid arthritis. Immunol Rev. 2001;184:184–202.
of RA, and there is scope for refinement of existing models to improve ani-
32. Kong YY, Feige U, Sarosi I, et al. Activated T cells regulate bone loss and joint destruction in
mal welfare. adjuvant arthritis through osteoprotegerin ligand. Nature. 1999;402(6759):304–309.
33. Pettit AR, Ji H, von Stechow D, et al. TRANCE/RANKL knockout mice are protected from
REFERENCES bone erosion in a serum transfer model of arthritis. Am J Pathol. 2001;159(5):1689–1699.
34. Cromartie WJ, Craddock JG, Schwab JH, Anderle SK, Yang CH. Arthritis in rats after sys-
1. Nabozny GH, Bull MJ, Hanson J, Griffiths MM, Luthra HS, David CS. Collagen-induced temic injection of streptococcal cells or cell walls. J Exp Med. 1977;146(6):1585–1602.
arthritis in T cell receptor Vβ congenic B10.Q mice. J Exp Med. 1994;180:517–524. 35. Kuiper S, Joosten LA, Bendele AM, et al. Different roles of tumour necrosis factor alpha and
2. Campbell IK, Hamilton JA, Wicks IP. Collagen-induced arthritis in C57BL/6 (H-2b) mice: interleukin 1 in murine streptococcal cell wall arthritis. Cytokine. 1998;10(9):690–702.
new insights into an important disease model of rheumatoid arthritis. Eur J Immunol. 36. Joosten LA, Abdollahi-Roodsaz S, Heuvelmans-Jacobs M, et al. T cell dependence of chronic
2000;30(6):1568–1575. destructive murine arthritis induced by repeated local activation of Toll-like receptor-driven

Descargado para Grecia Vera Vallejos (greciav.9@hotmail.com) en Ricardo Palma University de ClinicalKey.es por Elsevier en junio 15, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
804 SECTION 7  Rheumatoid Arthritis
pathways: crucial role of both interleukin-1beta and interleukin-17. Arthritis Rheum. 57. Cantaert T, Teitsma C, Tak PP, Baeten D. Presence and role of anti-citrullinated protein antibod-
2008;58(1):98–108. ies in experimental arthritis models. Arthritis Rheum. 2013;65(4):939–948.
37. Deng GM, Tarkowski A. Synovial cytokine mRNA expression during arthritis triggered by 58. Sakaguchi N, Takahashi T, Hata H, et al. Altered thymic T-cell selection due to a mutation of
CpG motifs of bacterial DNA. Arthritis Res. 2001;3(1):48–53. the ZAP-70 gene causes autoimmune arthritis in mice. Nature. 2003;426(6965):454–460.
38. Dumonde DC, Glynn LE. The production of arthritis in rabbits by an immunological reac- 59. Yoshitomi H, Sakaguchi N, Kobayashi K, et al. A role for fungal {beta}-glucans and their
tion to fibrin. Br J Exp Pathol. 1962;43:373–383. receptor Dectin-1 in the induction of autoimmune arthritis in genetically susceptible mice.
39. van den Berg WB, van de Putte LB, Zwarts WA, Joosten LA. Electrical charge of the antigen J Exp Med. 2005;201(6):949–960.
determines intraarticular antigen handling and chronicity of arthritis in mice. J Clin Invest. 60.
Atsumi T, Ishihara K, Kamimura D, et al. A point mutation of Tyr-759 in interleu-
1984;74(5):1850–1859. kin 6 family cytokine receptor subunit gp130 causes autoimmune arthritis. J Exp Med.
40. Cooke TD, Hurd ER, Ziff M, Jasin HE. The pathogenesis of chronic inflammation in experi- 2002;196(7):979–990.
mental antigen-induced arthritis. II. Preferential localization of antigen-antibody complexes 61. Keffer J, Probert L, Cazlaris H, et al. Transgenic mice expressing human tumour necrosis
to collagenous tissues. J Exp Med. 1972;135(2):323–338. factor: a predictive genetic model of arthritis. EMBO J. 1991;10:4025–4031.
41. Koenders MI, Lubberts E, Oppers-Walgreen B, et al. Blocking of interleukin-17 during reac- 62. Kontoyiannis D, Pasparakis M, Pizarro TT, Cominelli F, Kollias G. Impaired on/off regula-
tivation of experimental arthritis prevents joint inflammation and bone erosion by decreas- tion of TNF biosynthesis in mice lacking TNF AU-rich elements: implications for joint and
ing RANKL and interleukin-1. Am J Pathol. 2005;167(1):141–149. gut-associated immunopathologies. Immunity. 1999;10(3):387–398.
42. Abdollahi-Roodsaz S, Joosten LA, Helsen MM, et al. Shift from Toll-like receptor 2 (TLR- 63. Probert L, Plows D, Kontogeorgos G, Kollias G. The type I interleukin-1 receptor acts in
2) toward TLR-4 dependency in the erosive stage of chronic streptococcal cell wall series with tumor necrosis factor (TNF) to induce arthritis in TNF-transgenic mice. Eur J
arthritis coincident with TLR-4-mediated interleukin-17 production. Arthritis Rheum. Immunol. 1995;25:1794–1797.
2008;58(12):3753–3764. 64. Brennan FM, Chantry D, Jackson A, Maini R, Feldmann M. Inhibitory effect of TNFα antibod-
43. Jacobs JP, Wu HJ, Benoist C, Mathis D. IL-17-producing T cells can augment autoanti- ies on synovial cell interleukin-1 production in rheumatoid arthritis. Lancet. 1989;2:244–247.
body-induced arthritis. Proc Natl Acad Sci U S A. 2009;106(51):21789–21794. 65. Niki Y, Yamada H, Seki S, et al. Macrophage- and neutrophil-dominant arthritis in human
44. Kouskoff V, Korganow AS, Duchatelle V, Degott C, Benoist C, Mathis D. Organ-specific IL-1 alpha transgenic mice. J Clin Invest. 2001;107(9):1127–1135.
disease provoked by systemic autoimmunity. Cell. 1996;87(5):811–822. 66. Fell HB, Jubb RW. The effect of synovial tissue on the breakdown of articular cartilage in
45. Peccoud J, Dellabona P, Allen P, Benoist C, Mathis D. Delineation of antigen contact residues organ culture. Arthritis Rheum Sep. 1977;20(7):1359–1371.
on an MHC class II molecule. EMBO J. 1990;9(13):4215–4223. 67. Saklatvala J, Pilsworth LM, Sarsfield SJ, Gavrilovic J, Heath JK. Pig catabolin is a form
46. Korganow AS, Ji H, Mangialaio S, et al. From systemic T cell self-reactivity to organ-specific of interleukin 1. Cartilage and bone resorb, fibroblasts make prostaglandin and collage-
autoimmune disease via immunoglobulins. Immunity. 1999;10(4):451–461. nase, and thymocyte proliferation is augmented in response to one protein. Biochem J.
47. Mangialaio S, Ji H, Korganow AS, Kouskoff V, Benoist C, Mathis D. The arthritogenic 1984;224(2):461–466.
T cell receptor and its ligand in a model of spontaneous arthritis. Arthritis Rheum. 68. Saklatvala J, Sarsfield SJ, Townsend Y. Pig interleukin 1. Purification of two immunologi-
1999;42(12):2517–2523. cally different leukocyte proteins that cause cartilage resorption, lymphocyte activation, and
48. Solomon S, Kolb C, Mohanty S, et al. Transmission of antibody-induced arthritis is indepen- fever. J Exp Med. 1985;162(4):1208–1222.
dent of complement component 4 (C4) and the complement receptors 1 and 2 (CD21/35). 69. Horai R, Saijo S, Tanioka H, et al. Development of chronic inflammatory arthropathy resem-
Eur J Immunol. 2002;32(3):644–651. bling rheumatoid arthritis in interleukin 1 receptor antagonist-deficient mice. J Exp Med.
49. Corr M, Crain B. The role of FcgammaR signaling in the K/B x N serum transfer model of 2000;191(2):313–320.
arthritis. J Immunol. 2002;169(11):6604–6609. 70. Abdollahi-Roodsaz S, Joosten LA, Koenders MI, et al. Stimulation of TLR2 and TLR4
50. Maccioni M, Zeder-Lutz G, Huang H, et al. Arthritogenic monoclonal antibodies from K/ differentially skews the balance of T cells in a mouse model of arthritis. J Clin Invest.
BxN mice. J Exp Med. 2002;195(8):1071–1077. 2008;118(1):205–216.
51. Kyburz D, Carson DA, Corr M. The role of CD40 ligand and tumor necrosis factor alpha 71. Nakae S, Saijo S, Horai R, Sudo K, Mori S, Iwakura Y. IL-17 production from activated T
signaling in the transgenic K/BxN mouse model of rheumatoid arthritis. Arthritis Rheum. cells is required for the spontaneous development of destructive arthritis in mice deficient
2000;43(11):2571–2577. in IL-1 receptor antagonist. Proc Natl Acad Sci U S A. 2003;100(10):5986–5990.
52. Herve CA, Wait R, Venables PJ. Glucose-6-phosphate isomerase is not a specific autoantigen 72. Hawkins P, Armstrong R, Boden T, et al. Applying refinement to the use of mice and rats in
in rheumatoid arthritis. Rheumatology (Oxford). 2003;42(8):986–988. rheumatoid arthritis research. Inflammopharmacology. 2015;23(4):131–150.
53.
Matsumoto I, Lee DM, Goldbach-Mansky R, et al. Low prevalence of antibodies to 73. Trentham DE, Townes AS, Kang AH. Autoimmunity to type II collagen an experimental
glucose-6-phosphate isomerase in patients with rheumatoid arthritis and a spectrum of model of arthritis. J Exp Med. 1977;146:857–868.
other chronic autoimmune disorders. Arthritis Rheum. 2003;48(4):944–954. 74. Finnegan A, Mikecz K, Tao P, Glant TT. Proteoglycan (aggrecan)-induced arthritis in BALB/c
54. Schubert D, Maier B, Morawietz L, Krenn V, Kamradt T. Immunization with glucose-6-phos- mice is a Th1-type disease regulated by Th2 cytokines. J Immunol. 1999;163(10):5383–5390.
phate isomerase induces T cell-dependent peripheral polyarthritis in genetically unaltered 75. Wooley PH, Seibold JR, Whalen JD, Chapdelaine JM. Pristane-induced arthritis. The immu-
mice. J Immunol. 2004;172(7):4503–4509. nologic and genetic features of an experimental murine model of autoimmune disease.
55. Uysal H, Bockermann R, Nandakumar KS, et al. Structure and pathogenicity of anti- Arthritis Rheum. 1989;32:1022–1030.
bodies specific for citrullinated collagen type II in experimental arthritis. J Exp Med. 76. Vingsbo C, Sahlstrand P, Brun JG, Jonsson R, Saxne T, Holmdahl R. Pristane-induced arthri-
2009;206(2):449–462. tis in rats: a new model for rheumatoid arthritis with a chronic disease course influenced by
56. Maresz KJ, Hellvard A, Sroka A, et al. Porphyromonas gingivalis facilitates the development both major histocompatibility complex and non-major histocompatibility complex genes.
and progression of destructive arthritis through its unique bacterial peptidylarginine deiminase Am J Pathol. 1996;149(5):1675–1683.
(PAD). PLoS Pathog. 2013;9(9):e1003627.

Descargado para Grecia Vera Vallejos (greciav.9@hotmail.com) en Ricardo Palma University de ClinicalKey.es por Elsevier en junio 15, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.

You might also like