You are on page 1of 30

CHAPTER 56 

Autoantibodies in
Rheumatoid Arthritis
Felipe Andrade • Erika Darrah • Antony Rosen

KEY POINTS ifications (PTMs) as critical determinants targeted by auto-


antibodies in RA, which has led to the identification of
Autoantibodies in rheumatoid arthritis (RA) are important novel autoantibody systems and pathogenic mechanisms in
tools for diagnosis and for defining pathogenic pathways in
this disease (Table 56-1). In this chapter we will review the
this disease.
autoantibodies in RA, with emphasis on rheumatoid factors
Rheumatoid factors (RFs) are autoantibodies that recognize (RFs), anti-citrullinated protein autoantibodies (ACPAs),
the Fc portion of immunoglobulin G molecules. They were and the anti–peptidylarginine deaminase (PAD) autoanti-
the first autoantibodies described in RA. bodies. The implications of these specificities for diagnosis
RFs have numerous causes in addition to RA and therefore and prediction of outcome, as well as for understanding
have limited specificity for RA. pathogenic events in the disease, will be highlighted.
Autoantibodies targeting citrullinated proteins are hallmarks
of the immune response in RA. RHEUMATOID FACTOR
The anti–cyclic citrullinated peptide (anti-CCP) assay broadly
The earliest assays (later called the Rose-Waaler agglutina-
detects antibodies against anti-citrullinated protein
antibodies (ACPAs) and has high specificity and excellent
tion test) that suggested the existence of autoantibodies in
sensitivity in RA. RA were developed in the early 1940s, when sera from
patients with RA were shown to cause agglutination of
ACPAs precede the onset of clinical RA and are associated sheep blood cells that had been sensitized with sub-
with more severe disease, making them useful as diagnostic
agglutinating doses of rabbit anti–sheep erythrocyte anti-
and prognostic tools.
bodies.1,2 These assays were later shown to be detecting
Peptidylarginine deiminase enzymes, which catalyze the immunoglobulin (Ig)M antibodies from patients with RA
conversion of peptidyl-arginine to peptidyl-citrulline, play that recognized the Fc portion of IgG.3 Numerous modifica-
important roles in generating citrullinated autoantigens tions of the agglutination assay evolved, particularly the use
in RA.
of IgG-coated latex beads instead of sheep erythrocytes.4,5
Genetic factors such as “shared epitope” human leukocyte Subsequent developments established RF assays in radioim-
antigen–DR subregion alleles appear to interact with munoassay, enzyme-linked immunosorbent assay (ELISA),
environmental factors, such as smoking and infection, to and nephelometry formats, with increased convenience but
initiate and drive autoimmunity in RA. similar performance in terms of sensitivity and specificity.
RFs may be positive in healthy control subjects (1% in
younger persons and up to 5% in persons older than 70
Autoantibodies have proven to be very useful tools for diag- years) and in patients with numerous other non-RA diseases
nosis and prediction in autoimmune rheumatic diseases. (including other rheumatic diseases such as Sjögren’s syn-
Emerging data pertaining to several autoimmune rheumatic drome and cryoglobulinemia), as well as chronic infections.6-8
diseases have demonstrated that the clinical evolution of The pretest probability of diagnosing RA therefore greatly
disease from the pre-clinical phase to overt clinical disease influences the performance of the RF test, with sensitivities
is marked by a change in the specificity of the immune and specificities both in the 50% to 90% range, depending
response, with autoantibodies directed against distinct on the patient groups studied.9,10 The existence of IgG and
antigenic targets at different disease phases. Whereas IgA RFs and evidence of somatic hypermutation have
numerous autoimmune rheumatic diseases traditionally suggested that some RFs in RA are T cell dependent.11,12
have been marked by highly phenotype-specific autoanti- Although some studies have suggested that IgG and IgA
body responses (e.g., anti–double-stranded DNA antibodies RFs increase the specificity of RFs for RA,13 a recent meta-
in systemic lupus erythematosus [SLE] and high-titer anti– analysis showed that assays of the different RF isotypes per-
topoisomerase-1 antibodies in diffuse scleroderma), the formed very similarly in terms of sensitivity and specificity
discovery of highly specific autoantibody markers of rheu- and therefore do not add much compared with standard
matoid arthritis (RA) lagged significantly behind these RF assays.14
other diseases. During the past decade, enormous progress Importantly, patients with RA also vary in terms of
has occurred in this area, largely fueled by the discovery that timing of the appearance of RF, with RF preceding symp-
citrullinated proteins are specific targets of autoantibodies tomatic disease in a significant subpopulation15-18 and fol-
in RA. This finding has highlighted post-translational mod- lowing disease onset with variable kinetics in other patients.

831
832 PART 7    DIAGNOSTIC TESTS AND PROCEDURES IN RHEUMATIC DIEASES

TABLE 56-1  Native and Modified Targets of Autoantibodies in Rheumatoid Arthritis


Prevalence of
Type of Antigen Type of PTM Antigen Generation Protein Autoantibodies
Native None Unknown IgG (Fc) 50%-90%
hnRNP-A2 (RA33) 35%
G6PI 12%-29%
PAD4 30%-40%
Post-translationally Citrullination Enzymatic reaction mediated by Vimentin, fibronectin, actin, HSP90, Up to 80%
modified PADs; arginine residues are histones, α-enolase, eEF1a, CAP-1,
changed to citrulline CapZalpha-1, asporin, cathepsin D,
histamine receptor, PDI, ER60
precursor, ALDH2, collagen type I and
II, eIF4GI, aldolase, PGK1, calreticulin,
HSP60, FUSE-BP1 and 2, ApoE, MNDA
Carbamylation Nonenzymatic reaction of cyanate Unknown 45%
with lysine residues; lysine is
changed to homocitrulline
MAA adducts Nonenzymatic reaction; peroxidation Unknown 29%-93%
of membrane lipids generate
reactive aldehydes that modify
lysine residues, generating MAA
adducts

ALDH2, Mitochondrial aldehyde dehydrogenase; ApoE, apolipoprotein E; CAP1, adenylyl cyclase–associated protein-1; CapZalpha-1, F-actin capping
protein alpha-1 subunit; eEF1a, elongation factor 1-alpha; eIF4GI, eukaryotic translation initiation factor-4G1; FUSE-BP, far upstream element binding
protein; G6PI, glucose-6-phosphate isomerase; IgG, immunoglobulin G; hnRNP, heterogeneous nuclear ribonucleoprotein; HSP, heat shock protein; MAA,
malondialdehyde-acetaldehyde; MNDA, myeloid nuclear differentiation antigen; PAD, peptidylarginine deaminase; PDI, protein disulfide-isomerase; PGK1,
phosphoglycerate kinase-1; PTM, post-translational modification.

Indeed, the earlier onset of RF in patients with RA has been ings: (1) their lack of specificity for RA and (2) an unclear
associated with more severe disease, highlighting a possible kinetic and mechanistic connection to pathogenesis. Thus
contribution of these antibodies to amplification.19 The defining additional autoantibodies specific for RA has been
observation that RF only appears after disease onset in some a major priority. A significant discovery in this regard was
patients suggests that it may mark distinct, sequential events made initially by Nienhuis and Mandema in 196421 (Figure
in pathogenesis, which are very close together in the sub- 56-1). These investigators recognized an autoantibody that
group in whom RF occurs early, or are separated in time in stained keratohyalin granules surrounding the nucleus in
patients in whom RF follows symptoms. The mechanisms cells of human buccal mucosa, which they called antiperi-
that drive the production of RF in persons with RA are not nuclear factor (APF). These APF antibodies were found in
fully understood. The presence of somatic hypermutation in 49% to 91% of patients with RA,21-25 and they have reported
RFs and the evidence that these antibodies are produced by specificities of 73% to 99%.21,24-27 The assay was difficult to
plasma cells in the rheumatoid synovium have suggested standardize because not every donor of buccal mucosal cells
that the RF response is driven locally by antigen.11,12 demonstrated the staining pattern,28-31 and thus the assay
However, it is still unclear how the IgG becomes self- did not find its way into routine clinical practice, but the
immunogenic in the rheumatoid joint. In contrast to other specificity for RA suggested that the assay might be report-
autoantigens in RA, there is no evidence that the IgG ing on a potentially important pathogenic pathway. A
targeted by RFs requires modification (e.g., PTM) for anti- second research group subsequently demonstrated that RA
body recognition. Nevertheless, it is still possible that the sera recognized antigens in the stratified squamous layer of
initial event that breaks tolerance to IgG may be initiated esophageal epithelium—a staining pattern they termed
through an abnormally modified molecule, as occurs for antikeratin antibodies (AKAs).32 Subsequent iterations of
other autoantigens in RA. The possible mechanisms this assay utilized other forms of stratified squamous epithe-
whereby RF may play an amplifying role in RA are numer- lium, including human skin.33 Although this AKA assay
ous and include amplification of antigen capture, signaling, also had very good specificity properties, it was of limited
and effector functions, among others.20 sensitivity34-37 and was not particularly convenient or easy
to standardize. Thus neither assay was broadly used in the
clinical diagnostic arena. Like the LE cell in SLE, however,
ANTIBODIES AGAINST CITRULLINATED the assays provided a framework for antigen discovery that
ANTIGENS would later lead to highly specific and convenient tools for
RA diagnosis and classification.
Antikeratin Antibodies and Antiperinuclear
Factor: Initial Insights into a Novel Group of
Autoantibodies in Rheumatoid Arthritis Discovery of Autoantibodies That
Recognize Peptidylcitrulline
Although RFs have been clinically useful in diagnosis of
RA—particularly in cases in which the index of clinical The initial discovery of antibodies that recognized keratin-
suspicion is high—these autoantibodies have two shortcom- ized squamous epithelium in patients with RA prompted
CHAPTER 56    Autoantibodies in Rheumatoid Arthritis 833

4
1) 5
CP
(C

s
en
3
rin 2

ig
pe
d 21

d 32

nt
ts
gg

lo

in a
e
et
ie

ie

ve

jo uto
ila

rg
rg
tif

tif

of

de

ta
d 38

ta

RA of a
en

en

t
pr

s 59 e
A
be
id

id

ie

ie to b
th

IS

th tion
tif
)

wi

s 44 to
PF

KA

EL
en

ib nd

s fica
od nd
d
(A

(A

id

e
e
ize

u
id
tib fou

an fo
s

od
or

PA nti
s

ie

pt
ie
l

in
ie

ca
ct

rin rin

AC ide
od

pe
an in
od
fa

lo

t
F gr

gg fib
tib
tib

by he
ar

co

AP lag

e
an

fila /β
an
le

at

t
to

ti- d α
A/ d fi

ed in
uc

llin
rin
in

nd

rg ion
rin

AK te
at

an te
ru
gg
u

of llina
r

of ina
pe

cit

ta ns
ke

fo

fila

et
ti–

pa
l
c
ti-

l
ti-

tru

tru
cli
AP
An

An

Ex
An

Cy
Ci

Ci
1964 1979 1991 1993 1998 2000 2001 2004

Figure 56-1  A timeline of the discovery of anti-citrullinated protein antibodies (ACPAs) in rheumatoid arthritis (RA). ELISA, Enzyme-linked immuno-
sorbent assay.

studies to define the target of these antibodies. It was ini- filaggrin. It is important to highlight that antibodies against
tially shown that APF only stained a fully differentiated citrullinated proteins in RA do not recognize free citrulline
squamous epithelial layer and that APF staining was identi- but only citrulline residues (i.e., peptidylcitrullines) within
cal to the pattern produced by a monoclonal antibody spe- the context of peptides or protein sequences.
cific for human (pro)filaggrin in indirect immunofluorescence These initial studies became the basis for the major dis-
of permeabilized squamous epithelial cells.23 Although the covery that protein sequences containing citrulline residues
antigen could not be definitively identified using this are one of the most prominent targets for autoantibodies
co-localization alone, it suggested that the differentiation in RA. However, because the epidermis is not a target of
state might affect antigen recognition. Subsequently, RA rheumatoid inflammation and there is no evidence that
sera were shown to recognize a 40 kDa protein extracted (pro)filaggrin is expressed in articular tissues, this molecule
from human epidermis, which was identified as a neutral/ cannot be the autoantigen that drives the AKA/APF
acidic isoform of filaggrin.38 IgG from RA sera affinity- response in the joint. The fact that anti-citrullinated filag-
purified against the 40 kDa filaggrin protein was reactive in grin antibodies were enriched in synovial tissue compared
both the APF and AKA tests, demonstrating that these with the serum or synovial fluid45 and that these antibodies
autoantibodies were very similar or identical.39 are synthesized locally by plasmocytes within the rheuma-
Filaggrin (filament-aggregating protein) is produced toid pannus46 strongly suggests that anti-citrullinated filag-
during the late stages of terminal differentiation of epithe- grin cross-reacts with an antigen enriched in the RA joint/
lial cells. It is synthesized as a heavily phosphorylated pre- synovial tissue. Because citrullination is a very frequent
cursor protein (profilaggrin) that consists of 10 to 12 filaggrin event in different tissues,47-51 great caution must be exer-
repeats.40 Profilaggrin is deposited in granules, and filaggrin cised when attempting to ascribe a primary role for a specific
is released by proteolytic cleavage during differentiation of antigen in driving the antipeptidylcitrulline antibody
the cells by proteases that remain to be fully defined. Coin- response. The relevance of filaggrin in RA is therefore
cident with cleavage, the protein is dephosphorylated, and largely historic and unlikely to be of pathogenic relevance.
a significant proportion (~20%) of the arginine residues are Instead, its conformation and high content of citrulline
deaminated (i.e., converted to citrulline),41,42 a PTM medi- residues make it an excellent surrogate with which to detect
ated by the PAD enzymes (see later section Citrullinated antibodies against citrullinated molecules. Many other
Antigen Generation in Rheumatoid Arthritis).43 Because citrullinated autoantigens found in the rheumatoid joint
RA sera appeared to specifically target the neutral isoform that are currently being characterized are much more likely
of filaggrin (which is present and citrullinated in fully dif- to be of pathogenic relevance (see later section Anti-
ferentiated squamous epithelia), van Venrooij and col- citrullinated Protein Antibodies).
leagues addressed whether RA sera that were positive in the
AKA/APF assays specifically recognized citrullinated pep- Anti–Cyclic Citrullinated Peptide Antibodies
tides.44 Thus regions within the deduced amino acid
sequence of human profilaggrin with a high antigenicity In the initial characterization of AKA/APF antibodies, van
index and the largest number of arginine residues were Venrooij and colleagues used a single C-terminal peptide
selected to generate synthetic peptides where arginine resi- derived from filaggrin (amino acids 306 to 324) to generate
dues were substituted with citrulline. Using these citrulli- nine variants in which five arginine residues were changed
nated peptides, van Venrooij and colleagues demonstrated to citrullines, either individually or in pairs, and these pep-
that AKA/APF antibodies are directed against citrullinated tides were all assayed by ELISA.44 Interestingly, although
834 PART 7    DIAGNOSTIC TESTS AND PROCEDURES IN RHEUMATIC DIEASES

the peptides were almost identical (the only difference being Using protein extracts from rheumatoid synovial mem-
that the citrulline residues had different positions within branes and affinity purified anti-citrullinated filaggrin anti-
the peptide), remarkable differences were found in the serum bodies, Masson-Bessière and colleagues showed that RA
reactivity patterns toward each peptide (from 20% to 48% synovial tissue contains several citrullinated proteins and
positivity), suggesting that although citrullination plays a defined the α and β chains of fibrin(ogen) as the first local
critical role in antigen recognition by AKA/APF antibodies, targets for ACPAs in RA synovial tissue.59 Because circulat-
the modification per se is not the only determinant that ing fibrinogen is not citrullinated, the presence of citrulli-
confers antibody binding. Instead, the data suggested that nated fibrin in the rheumatoid synovium strongly suggested
AKA/APF represent a pool of antibodies that recognize that after fibrin deposition, citrullination occurs in situ
citrulline residues depending on the context of their sur- through the activity of locally expressed PAD. This proposal
rounding amino acids. Indeed, when data from all peptides (i.e., that the joint is the site for autoantigen citrullination
were pooled, the sensitivity increased to as much as 76%.44 in RA) has been further supported and extended to many
Because peptides often adopt a β-turn conformation other citrullinated autoantigens in RA.60
within the antibody–peptide complex52 and cysteine- To date, several different approaches have been used to
bridged cyclic peptides have been shown to mimic the β- identify potential ACPA targets in RA. Although these
turn structure of the original antigenic determinant and can approaches all utilize protein sequencing to identify auto-
bind with enhanced affinity to antibodies,53 van Venrooij antigens detected by ACPAs using immunoblotting assays,
and colleagues engineered a cyclic peptide (substituting the they vary in terms of the source of the antigens. Thus, in
terminal serine residues with cysteines and cyclizing the addition to RA pannus, identification of citrullinated auto-
peptide through the formation of a disulfide bond) to which antigens have been extended to rheumatoid synovial fluid,
anti–citrullinated peptide antibodies reacted with higher as well as to cell lysates in which purified PADs have been
affinity.54 Using this cyclic citrullinated peptide (later used to generate in vitro citrullinated proteins.
named CCP1) and its linear counterpart, it was demon- Using these approaches, several citrullinated autoanti-
strated that the cyclic structure increased the sensitivity of gen candidates have also been identified. These candidates
the assay (68% vs. 49%, without affecting specificity),44,54 include vimentin61,62; fibronectin63; actin64; heat shock
although it was still less sensitive than the assays using the protein (HSP)9065; histones66; α-enolase, elongation factor-
combination of linear peptides (i.e., 76% vs. 68%).44 Nev- 1α, and adenylyl cyclase–associated protein-167; F-actin
ertheless, CCP1 became the antigen for the first generation capping protein α-1 subunit, asporin, cathepsin D, hista-
of ELISAs designed to detect antibodies against citrulli- mine receptor, protein disulfide-isomerase, ER60 precursor,
nated autoantigens. and mitochondrial aldehyde dehydrogenase68; collagen type
To improve the CCP1 test, libraries of citrullinated pep- I69 and II70; eukaryotic translation initiation factor 4G171;
tides were used to construct the second-generation anti- aldolase, phosphoglycerate kinase-1 (PGK1), calreticulin,
CCP assay (CCP2), which was broadly adopted for clinical HSP60, and the far upstream element binding proteins
use.55,56 In 2005, a third generation of anti–cyclic citrulli- (FUSE-BP) 1 and 272; and apolipoprotein E and myeloid
nated peptide (CCP3) was made available for the laboratory nuclear differentiation antigen.73 Interestingly, some of
diagnosis of RA. These assays have been reported to recog- these molecules (e.g., vimentin, α-enolase, collagen type II,
nize additional citrullinated epitopes that are not identifi- HSP60, aldolase, and calreticulin) had been previously
able with the second-generation CCP assays. In studies identified as autoantigens in RA, before it was recognized
directly comparing second- and third-generation anti-CCP that protein citrullination played a role in their recognition
assays, the assays performed very similarly,57,58 with a slightly by autoantibodies.67,74 Although RA sera containing ACPAs
increased sensitivity of CCP3 in some studies (e.g., sensi- may recognize both the native and citrullinated forms of the
tivities of 82.9% vs. 78.6% for CCP2, with specificities in antigen, the modified form is usually preferentially recog-
the 93% to 94% range).58 nized. Although this group of citrullinated antigens repre-
sents proteins that are recognized by RA antibodies, the
ANTI-CITRULLINATED PROTEIN number of citrullinated proteins found in the rheumatoid
ANTIBODIES joint is more extensive and includes several dozen molecules
that have not yet been characterized.73,75,76 Whether unique
The finding that autoantibodies in RA recognize peptide ACPAs exist for each one of these molecules or whether
sequences containing citrulline residues and that filaggrin only a few citrullinated autoantigens are responsible for
was unlikely to be the physiologic target of these antibodies driving the complete ACPA response in RA is still unknown.
prompted the search for the primary protein targets of the Among the citrullinated candidate antigens present in
antibodies detected by the CCP assay. It was believed that the joint, the best characterized clinically and pathogeni-
the identification of these antigens may provide additional cally are fibrin(ogen), vimentin, collagen type II, and
information with regard to patient subtyping (compared α-enolase. ELISA-based assays have been developed using
with the general anti-CCP assay), as well as novel insights citrullinated proteins (e.g., fibrinogen) or citrullinated pep-
into disease etiology and pathogenesis. With the initial dis- tides (e.g., vimentin, collagen type II, and α-enolase)
covery of some of these citrullinated antigens, the name derived from these putative RA autoantigens to detect
anti-citrullinated protein antibodies (ACPAs) was introduced ACPAs. In the case of vimentin, the citrullinated sequence
to refer to RA autoantibodies for which detection was per- from a mutated isoform isolated from RA synovial fluid
formed using citrullinated proteins/peptides from putative (named mutated citrullinated vimentin [MCV]) has been
RA-associated autoantigens instead of filaggrin-derived or more commonly used instead of the native sequence.77
commercial CCPs. Although it was greatly anticipated that the detection of
CHAPTER 56    Autoantibodies in Rheumatoid Arthritis 835

ACPAs targeting single antigens would provide additional 1. Anti-CCP antibodies have high specificity for the diagnosis of
diagnostic information compared with the anti-CCP ELISA, RA. In systematic reviews and meta-analyses, anti-CCP
studies in this regard are inconclusive. Thus, in the absence antibody positivity has been shown to be as sensitive as
of convincing evidence that measuring individual ACPAs but more specific than RF in distinguishing RA from
provides any additional information compared with the other forms of inflammatory arthritis.14
anti-CCP assay, it appears that the use of CCPs is still a 2. The presence of anti-CCP autoantibodies is an important
more reliable test to confirm the presence of antibodies predictor of development of RA. RA developed within 3
against citrullinated proteins. In the case of patients who years in more than 90% of the patients with undifferenti-
are anti-CCP negative, the use of antigen-specific assays ated arthritis who tested positive for anti-CCP, in con-
may provide an additional tool to detect ACPAs. trast to only 25% of the patients who tested negative for
Because single ACPAs appear to have limited utility in anti-CCP.80
RA, further assays have been introduced to measure more 3. Anti-CCP positivity has been associated with a more severe
than one ACPA simultaneously. For example, autoantibod- and destructive disease course, although the independent role
ies targeting putative RA-associated citrullinated autoanti- of the antibody in comparison with RF has not yet been
gens are measured using custom Bio-Plex bead-based confirmed. Some investigators have shown that both RF
autoantibody assays (Bio-Rad Laboratories, Hercules, Calif.) and anti-CCP antibodies are independent predictors of
in which antigens (usually peptide sequences) are conju- severity, whereas others have shown that anti-CCP anti-
gated to spectrally distinct beads.78 Autoantibody binding bodies rather than RF are the better predictor of radio-
to antigen-coated beads is detected with anti–human graphic progression,81-88 particularly in patients who were
phycoerythrin–labeled antibody and analysis with a Lu- seronegative for RF.88
minex 200 instrument (Luminex, Austin, Tex.). This assay 4. Anti-CCP/ACPAs are associated with RA-related interstitial
is slightly more sensitive than the CCP ELISA, detecting lung disease (ILD) and cardiovascular disease (CVD).89-95
ACPAs in approximately 10% of patients negative for anti- ILD and CVD are extra-articular manifestations with
CCP antibodies (representing 3.7% of the total patient high mortality in persons with RA. Although the mech-
cohort),79 a difference that might be explained by the array anistic significance of these associations is still unclear,
of peptides used on the assay. In addition, the bead-based one possibility is that these antibodies are markers of
multiplex assay has been useful to define the epitope spread- systemic inflammation that contributes to lung and car-
ing of ACPAs in pre-clinical RA.78 However, although the diovascular damage. Alternatively, it is noteworthy that
multiplex assay provides additional information with regard citrullination has been found in lung, myocardium, and
to unique ACPA specificities present in the patient, know- atherosclerotic plaque,51,96-98 suggesting the possibility
ing these specificities has not yet demonstrated any advan- that these antibodies are directly pathogenic by targeting
tages for diagnosis or prognosis in RA. extra-articular tissues containing citrullinated antigens.
The presence of anti-CCP antibodies in patients who
have ILD but do not have RA is intriguing and has
Anti–Cyclic Citrullinated Peptide Antibodies/
focused attention on the lung as a possible extra-articular
Anti-citrullinated Protein Antibodies:
site where RA may be initiated99,100 (see section Poten-
Clinical Relevance
tial Environmental Factors in Rheumatoid Arthritis
The clinical importance of autoantibodies against citrulli- Pathogenesis).
nated antigens in RA stems from the following favorable The early and accurate diagnosis of RA, together with
features (Figure 56-2): effective treatment with disease-modifying anti-rheumatic

PADs convert peptidylarginine


to citrulline

Anti-PAD4 and ACPAs Citrullinated proteins accumulate


predict disease severity in the target tissue together
with IgG and complement

Citrullination is induced in
Anti-PAD4 and ACPAs
PAD-expressing cells during
precede disease onset
activaton/damage

PAD4 polymorphisms are associated


PADs can autocitrullinate
with RA and anti-PAD4 antibodies
in some cohorts
A subtype of anti-PAD4 antibodies
enhance PAD4 activity
Figure 56-2  Protein citrullination as a hub in rheumatoid arthritis (RA) pathogenesis. ACPAs, Anti-citrullinated protein antibodies; PAD, peptidylar-
ginine deiminase.
836 PART 7    DIAGNOSTIC TESTS AND PROCEDURES IN RHEUMATIC DIEASES

drugs, decreases accrual of joint damage in RA. Because known for decades, the relationship between genetics and
anti-CCP/ACPAs are markers of disease severity and are the development of RA autoantibodies is just beginning to
detectable early in the disease course, they are powerful be elucidated.114 A subset of HLA-DR alleles termed the
tools to classify patients with early stage inflammatory “shared epitope” (SE) alleles includes HLA-DR*0101,
arthritis who will benefit from treatment.101-103 *0102, *0401, *0404, *0405, *0408, *0410, *1001, and
*1402 and is united by a conserved sequence of amino acids
(QRRAA, QKRAA, or RRRAA) on the alpha helix of
Kinetics of Appearance of Anti–Cyclic
the DR-β chain peptide-binding groove.115 SE alleles are
Citrullinated Peptide Antibodies/
strongly associated with the development of anti-CCP/
Anti-citrullinated Protein Antibodies in
ACPA but are not independently associated with RF.80 Fur-
Rheumatoid Arthritis
thermore, there appears to be a gene dosage effect on the
Recent studies have demonstrated that autoantibodies may relative risk of anti-CCP development with an OR of 3.3
precede the onset of clinical disease in both tissue-specific to 4.7 for patients with one SE allele and an OR of 11.8 to
and systemic autoimmune diseases.17,104-110 Defining events 13.3 for patients with two SE alleles.80,116,117 Analysis of
prior to clinical onset is challenging, and several different individual SE alleles has revealed that anti-CCP/ACPAs
study designs have been used to define the appearance of are predominantly associated with HLA-DR4 rather than
autoantibodies and their relationship to disease. The two HLA-DR1 SE alleles.118 Interestingly, smoking has been
general categories include (1) stored blood samples col- shown to skew the genetic association of SE alleles with
lected prior to disease onset (blood banks or military anti-CCP development, with HLA-DR1 and HLA-DR10
cohorts) and (2) prospective studies examining emergence SE alleles being more important in this patient group (see
of autoantibodies and disease in high-risk persons (often Anti-citrullinated Protein Antibodies in Rheumatoid Ar-
relatives of affected individuals).111 In RA, the former thritis: Insights into Disease Mechanism section).119
approach has clearly shown that autoantibodies precede RA In addition to SE alleles, a single nucleotide polymor-
diagnosis in many persons in whom seropositive RA subse- phism in the PTPN22 gene (1858C/T) is also associated
quently develops, often by 2 to 6 years.109,110,112,113 In several with anti-CCP antibodies with an OR of 3.80.120 PTPN22
studies, 20% to 60% of patients with RA were RF positive encodes for lymphoid protein tyrosine phosphatase and has
prior to diagnosis, and 30% to 60% of patients were positive been shown to be associated with several autoimmune dis-
for anti-CCP.109,110,112,113 With one exception,113 the preva- orders, including RA.121 The combination of PTPN22
lence of anti-CCP prior to diagnosis was almost twice as 1858C/T genotype and anti-CCP antibodies was 100% spe-
high as RF. Interestingly, whereas RF remained present con- cific for RA and confers a relative risk of 130.03. The
sistently, anti-CCP could vary over time, even disappearing 1858C/T polymorphism was not associated with RF isotypes
in some persons. In one study, pre-clinical anti-CCP posi- and appeared to act independently of SE alleles.120 Genetic
tivity was strongly associated with the development of factors such as the SE and PTPN22 may therefore play a
erosive RA (odds ratio [OR], 4.64; 95% confidence interval, role in the pre-clinical phase of RA, predisposing individu-
1.71 to 12.63; P < 0.01), whereas RF was not (P = 0.60).113 als to the generation of anti-CCP antibodies and suscepti-
The evolution of autoantibody epitope spreading in pre- bility to the overt disease phenotype.
clinical RA has been addressed using a multiplex assay. The
study confirmed that ACPA positivity predicts progression
to RA in asymptomatic persons and showed that the number Citrullinated Antigen Generation in
of ACPA specificities accumulate over time during the pre- Rheumatoid Arthritis
clinical phase of the disease.78 Moreover, the expansion of The Peptidylarginine Deiminase Enzymes
the ACPA response precedes the elevation of many inflam-
matory cytokines, including tumor necrosis factor (TNF), Citrulline is not part of the natural pool of amino acids used
interleukin (IL)-6, IL-12p70, and interferon (IFN)-γ, sug- for protein synthesis, and therefore citrulline residues need
gesting that ACPAs may be part of the driving inflammatory to be generated post-translationally once the protein has
process in RA.78 been synthesized. This PTM, termed arginine deamination or
Together, these data highlight that the development of citrullination, is mediated by the calcium-dependent PAD
autoimmunity against citrullinated antigens is often asymp- enzymes. PADs target arginine residues in proteins and
tomatic and generally precedes the onset of clinical disease. mediate hydrolysis of the arginine guanidinium side chain,
This presymptomatic phase is potentially of great impor- resulting in the generation of citrulline residues and
tance, because it may identify persons in whom important ammonia (Figure 56-3A). The PADs belong to a larger
precursor conditions for the subsequent development of RA group of guanidino-modifying enzymes called the ami­
have been satisfied. The subsequent events that convert this dinotransferase (AT) superfamily.122,123 This superfamily of
RA precursor into a chronic, self-sustaining process are not enzymes is expressed both in prokaryotes and eukaryotes
yet known, but defining them is of great importance. and includes the arginine deaminases, the dimethylarginine
dimethylaminohydrolases, and the dihydrolases.123 PADs
are found in numerous species from bacteria to humans.
Genetic Associations with Anti–Cyclic
Interestingly, the bacterial PADs are not dependent on
Citrullinated Peptide Antibodies/
calcium for activity and only contain an approximately
Anti-citrullinated Protein Antibodies
40 kDa catalytic domain, whereas vertebrate mammals
Although the association of specific human leukocyte have larger multidomain enzymes (~75 kDa) whose activity
antigen–DR region (HLA-DR) alleles and RA has been is regulated by calcium. Five PAD enzymes have been
CHAPTER 56    Autoantibodies in Rheumatoid Arthritis 837

Citrullination

PAD + Ca2+

H2N Protein Protein


O
+ NH H2O NH3 NH
NH2 NH2
A Peptidylarginine Peptidylcitrulline

Carbamylation

Cyanate
Protein Protein
+ NH3 NH
NH2
O
B Peptidyllysine Peptidylhomocitrulline

MAA adducts
Acetaldehyde (AA) FAAB MDHDC
+ H
Malondialdehyde (MDA) O
H
+ NH3 O NH CH3 N
Protein H Protein Protein
O CH3 O
H H
C Primary amine Malondialdehyde acetaldehyde adducts (MAA)
Figure 56-3  Post-translational modifications targeted by antibodies in rheumatoid arthritis (RA). A, During citrullination, the calcium-dependent
peptidylarginine deiminase (PAD) enzymes target arginine residues in proteins, resulting in the generation of citrulline residues and ammonia.
B, Carbamylation results from the reaction of cyanate with the primary amine group of peptidyllysine that is converted to peptidylhomocitrulline.
C, Malondialdehyde-acetaldehyde (MAA) adducts (FAAB and MDHDC) are generated from the reaction of malondialdehyde (MDA) and acetaldehyde
(AA) with primary amine groups of amino acid residues, preferentially peptidyllysine. FAAB, 2-formyl-3-(alkylamino)butanal; MDHDC, 4-methyl-1,
4-dihydropyridine-3,5-dicarbaldehyde.

identified in mammals, and the PADI genes are located at contact between the N-terminal domain of one molecule
a single cluster on chromosome 1p36.1.43,124 For historical and the C-terminal domain of the second. The N-terminal
reasons, these isozymes have been designated PAD1 to PAD4 domain includes two immunoglobulin-like subdomains
and PAD6. Human PAD4 was initially named PAD5 but (named subdomain 1 and 2) that are proposed to mediate
was later renamed PAD4 to reflect the fact that it is a true protein-protein interactions and/or substrate targeting.128
ortholog of this isoform.125 Although initial evidence Five and six Ca2+-binding sites were identified in the struc-
suggested that PAD4 was the only nuclear PAD,126 recent ture of PAD4 and PAD2, respectively, with Ca2+ binding
evidence suggests that some PAD2 may also reside in the inducing conformational changes required to generate the
nucleus.127 active site cleft. Interestingly, the calcium binding residues
The PADs are highly conserved and share 50% to 55% are highly conserved among PADs, except for PAD6, which
sequence identity with each other,128 with different PADs lacks some of these sites; the catalytic cysteine is also dif-
having distinct substrate preferences and expression in spe- ferent in PAD6, and together these findings have suggested
cific tissues. PAD1 is primarily expressed in uterus and skin. that PAD6 is not a deaminating enzyme.
PAD2 is more widely expressed in muscle, skin, brain, PAD1 to PAD4 rely strongly on the presence of Ca2+ for
spleen, secretory glands, monocytes, and neutrophils. PAD3 activity and require millimolar amounts of calcium (~2
is expressed in skin and neutrophils, PAD4 is expressed in to 5 millimolar) to be activated in vitro.136 The resting
hematopoietic cells (e.g., neutrophils and monocytes), and concentration of calcium in the cytoplasm is normally
PAD6 is broadly expressed in germ cells, peripheral blood maintained in the range of 50 to 100 nM and only rises to
leukocytes, lung, small intestine, liver, spleen, and skeletal 200 to 800 nM upon cell activation with physiologic
muscle.43,61,124,125,129-131 Because of their prominent expres- stimuli.137-140 The large discrepancy between the in vitro and
sion in rheumatoid synovial tissue and fluid,60,132,133 PAD2 cellular calcium requirements for PAD activation suggests
and PAD4 have gained prominence as potential candidates that additional mechanisms, such as allosteric effects of
that drive citrullination of self-antigens in RA. PTMs or partner binding, may regulate PAD activity in vivo
by modifying the calcium requirement of the enzyme.136 It
is also important to note that whereas PAD activation pro-
Peptidylarginine Deiminase Structure, Activity,
vides one level of regulation, the activity of the PADs may
and Regulation
also be negatively regulated through autocitrullination.
The three-dimensional structure of PAD4128,134 and PAD2135 This is not dissimilar to other enzymes, where automodifica-
has been solved and is likely representative of the broader tion (e.g., autophosphorylation) modifies enzymatic func-
family. PAD2 and PAD4 are dimers formed by head-to-tail tion. Autocitrullination has been described for PADs 1, 2,
838 PART 7    DIAGNOSTIC TESTS AND PROCEDURES IN RHEUMATIC DIEASES

3, and 4.141,142 In the case of PAD4, direct citrullination of cytoplasmic lattices, and female mice deficient in PAD6 are
arginines surrounding the active site cleft appears to have infertile but otherwise normal.131 The function of PAD6 in
major impact on activity and substrate binding.141 Besides other organs/tissues besides ovaries is unknown.
cell activation, it has been proposed that PADs may also be
activated under conditions where intra-cellular concentra- Peptidylarginine Deaminases in Rheumatoid Arthritis
tions of calcium approach extra-cellular concentrations (~1
to 1.4 millimolar), which may occur in dying cells experi- Multiple PADs are likely responsible for autoantigen citrul-
encing secondary necrosis or as result of membranolytic lination in RA, particularly PADs 2 and 4.60,132,133 Although
damage.75 PAD4 has some unique characteristics that have focused
significant attention on this protein, it is important to be
aware that no evidence exists to suggest that pathologic
Structural and Functional Implications
citrullination in RA is exclusively or preferentially medi-
of Protein Citrullination
ated by this isoform.
Arginine residues play a central role in the maintenance of It is of interest that several polymorphisms in PAD4
secondary and tertiary protein structure. The positively have been genetically associated with RA development,
charged guanidino group allows the formation of intramo- particularly in Asian populations.157-160 In this regard, two
lecular hydrogen bonds to backbone carbonyl oxygens and common haplotypes of the PADI4 gene were initially iden-
also intermolecularly between different proteins.143 During tified157 and designated “susceptible” or “nonsusceptible”
the process of citrullination, the net charge of the protein based on their relative frequency in patients with RA versus
is reduced by the loss of a positive charge per citrulline control subjects. In the initial population, the OR for the
residue, producing changes in protein structure and affect- susceptible haplotype was almost 2; this effect was not
ing intramolecular and intermolecular interactions.144 observed in most white populations. Changes in messenger
It is likely that these structural changes also have impor- RNA stability of the susceptible PADI4 reported in the
tant functional implications. Indeed, deamination has been initial paper were not associated with changes in enzyme
implicated in several physiologic processes. In the skin level. Furthermore, although the susceptible haplotype
(which expresses PADs 1, 2, and 348,130,145), deamination of generates a PAD4 molecule containing three amino acid
filaggrin (a component of the cornified cell envelope) is substitutions in the N-terminal region (i.e., Gly55-Ser,
critical for its degradation into free amino acids, which act Val82-Ala, and Gly112-Ala), current evidence suggests
as a natural moisturizing factor in the stratum corneum.48,145 that these changes have no effect on the function of the
Interestingly, PAD2-deficient mice have normal develop- protein.141,161 Indeed, they appear to affect conformation
ment and show no abnormalities in the skin,146 suggesting only within the N-terminal domain but not the active site
functional redundancy in mouse development. Whether located in the C-terminal domain.161 The fact that PAD4
PAD2 functions in the skin during injury or environmental acts as a head-to-tail dimer may nevertheless allow confor-
perturbations is currently unknown. In the immune system, mational differences in the N-terminus of one molecule to
PAD2 (and/or potentially other PADs) citrullinates chemo- influence the C-terminal domain of another and influence
kines, a process that modulates chemokine activity.147-150 catalysis or even inhibition by autocitrullination. It has
Thus PAD2 may play important roles in controlling effector been proposed that the PAD4 genotype may exert its effects
functions related to environmental triggers. on RA disease susceptibility as a consequence of its immu-
Human PAD4 (first called PAD5) was first found in nogenicity, rather than its enzyme activity.141 Consistent
human myeloid leukemia HL-60 cells induced to differenti- with this hypothesis is the observation that, in addition to
ate into granulocytes by retinoic acid125 and later described its ability to citrullinate RA autoantigens, PAD4 itself is an
in peripheral blood granulocytes.129 Histones H2A, H3, and RA autoantigen (see next section Anti–peptidylarginine
H4 and nucleophosmin/B23 were the first PAD4 substrates deiminase autoantibodies).162,163 Moreover, it is noteworthy
to be identified.151 PAD4 appears to function as a transcrip- that the PADI4 susceptible gene appears to contribute to
tional co-regulator, mediated by its ability to catalyze the the development and progression of RA regardless of ACPA
deamination of specific residues present in the N-terminal status,164,165 supporting the notion that the snp variant may
tails of histones.152-154 This function, together with the drive susceptibility through other mechanisms besides the
finding that PAD4 is expressed during granulocyte differen- production of citrullinated autoantigens.
tiation, initially suggested that this enzyme may have a role
during granulopoiesis. However, mice deficient in PAD4 ANTI–PEPTIDYLARGININE DEIMINASE
have normal development,155 suggesting that this enzyme AUTOANTIBODIES
has no essential roles in steady-state cellular functions,
granulopoiesis, or other developmental processes. Recent In addition to its prominent enzymatic role in the genera-
studies have demonstrated that PAD4-mediated citrullina- tion of citrullinated autoantigens, PAD4 itself is targeted by
tion of histones is required for neutrophil extra-cellular trap autoantibodies in a subset of patients with RA.162,163 These
(NET) formation and bacterial clearance,154,155 suggesting a antibodies recognize the unmodified form of PAD4, but it
role for this enzyme in immune and inflammatory effector has been suggested that autocitrullinated PAD4 may be
functions. more immunogenic in some persons.141 As has been observed
PAD6, the most recently defined member of the PAD for anti-CCP and RF, anti-PAD4 antibodies are frequently
family, was initially cloned from mouse oocytes and named present prior to the onset of any disease symptoms.107 Inter-
egg PAD (ePAD),156 although the enzyme is widely expressed estingly, in a cohort of white patients with RA who had
in different tissues. PAD6 is essential for the formation of established disease, the susceptible PAD4 haplotype was
CHAPTER 56    Autoantibodies in Rheumatoid Arthritis 839

strikingly associated with PAD4 autoantibodies, even tion. Alternatively, it has been proposed that PADs may be
though the association of the susceptible haplotype with released during NET formation, contributing to extra-
RA development has not been observed in white popula- cellular citrullination. Although NETs may be an important
tions.163 The sensitivity of PAD4 antibodies for RA is in the source of inflammatory signals and some citrullinated auto-
30% to 40% range, with a specificity of greater than 95%. antigens, the contribution of this process to generating the
PAD4 autoantibodies are associated with anti-CCP anti- whole set of citrullinated molecules found in the rheuma-
bodies and are independently associated with more severe, toid joint (i.e., the RA citrullinome) is unclear.170 Instead,
erosive RA that persists despite treatment with TNF inhibi- recent studies have found that immune pathways that form
tors.166,167 Recently, a subset of anti-PAD4 antibodies identi- pores in cell membranes and that are active in the RA joint
fied by their cross-reactivity to the related protein PAD3 (e.g., the complement system’s membrane attack complex
has been identified and appears to be the major driver [MAC] and perforin released from cytotoxic cells) are
of the previously seen associations with erosive disease potent activators of PADs and efficient inducers of the RA
severity and progression despite treatment.136 These anti- joint citrullinome.75 The membranolytic damage induced by
bodies were also associated with anti-CCP antibodies, as these pathways is associated with high intra-cellular calcium
well as the SE alleles. Furthermore, patients with PAD3/ flux,172 which likely explains the hyperactivation of PADs
PAD4 cross-reactive antibodies are at the highest risk for in the damaged cell. It is also possible that other forms of
the development of RA-associated ILD, an effect signifi- cell death may play a role in mediating enhanced citrullina-
cantly augmented in patients who were past or present tion in the RA joint, particularly if these death pathways
cigarette smokers (OR, 61.4).168 The causal relationship (e.g., pyroptosis or necroptosis) result in extra-cellular
between PAD3/PAD4 antibody development and cigarette leakage of PADs. The discovery that fibrinogen is heavily
smoking is unknown but suggests that environmental factors citrullinated in the RA joint provided initial evidence that
such as cigarette smoke may trigger the development of citrullination can indeed occur extra-cellularly.59 Further-
RA-associated autoantibodies in susceptible persons (see more, soluble PAD2 and PAD4 have been detected in the
later section Potential Environmental Factors in Rheuma- synovial fluid from patients with RA,60,173 suggesting that
toid Arthritis Pathogenesis). these enzymes can gain access to the extra-cellular space
during inflammation.
MECHANISMS OF CITRULLINATED Although the extra-cellular calcium concentration in
AUTOANTIGEN PRODUCTION IN synovial fluid (0.5 to 1 mM) is more than 1000-fold higher
RHEUMATOID ARTHRITIS than intra-cellular levels, it is still not optimal for maximal
PAD activity in vitro. Recent studies have demonstrated
Two potential mechanisms have been implicated in aber- that anti-PAD3/PAD4 cross-reactive autoantibodies are
rant PAD activation and the production of citrullinated able to enhance PAD4 activity at calcium concentrations
autoantigens in the rheumatoid joint. One mechanism found extra-cellularly.136 These anti-PAD3/PAD4 autoanti-
involves the hyperactivation of PADs intra-cellularly, and bodies decrease the enzymatic requirement for calcium into
the second relies on extra-cellular autoantigen citrullina- the physiologic range and greatly augment PAD4 activity.
tion. Importantly, these mechanisms are not mutually Thus PAD4 released by neutrophils during NET formation
exclusive but are complementary. Initial immunohisto- or other forms of cell deaths may encounter anti-PAD4
chemical studies of synovial tissue focused attention on antibodies in the synovial fluid with the capacity to activate
synoviocytes and inflammatory cells as potential sources of enzymatic function, contributing to extra-cellular citrulli-
PADs and citrullinated antigens in the RA joint.132,133 How- nated autoantigen generation in RA.
ever, whereas functional studies to address PAD expression,
activity, and production of citrullinated autoantigens in iso-
lated synoviocytes are limited and inconclusive,169 the study NOVEL POST-TRANSLATIONAL
of neutrophils has provided important insights into mecha- MODIFICATIONS TARGETED BY
nisms of autoantigen citrullination in RA.64,75,170 These cells ANTIBODIES IN RHEUMATOID ARTHRITIS
represent one of the most abundant cell types in RA syno- Anti–Carbamylated Protein Antibodies
vial fluid and constitutively express several PADs.64 Further-
more, they can hypercitrullinate and generate citrullinated Proteins that contain homocitrulline (i.e., carbamylated
autoantigens when activated with calcium ionophores,64 proteins) recently have been recognized as an important
and they are highly enriched in citrullinated autoantigens target of autoantibodies in RA, with IgG and IgA antibod-
when freshly isolated from the RA joint.75 Two mechanisms ies recognizing carbamylated antigens identified in more
have been proposed to trigger PAD activation and citrulli- than 40% of patients with RA.174 In contrast to citrul­
nated autoantigen production in neutrophils in RA: (1) lination, carbamylation is a nonenzymatic PTM in which
formation of neutrophil extra-cellular traps (NETs), an anti- homocitrulline is generated by the reaction of cyanate with
microbial system in which granule proteins and chromatin the primary amine of lysine residues (Figure 56-3B). Carba-
are extruded from the cell and form extra-cellular fibers that mylation therefore accumulates in conditions that enhance
bind pathogens,170 and (2) membranolytic damage mediated cyanate levels, such as uremia, inflammation, and cigarette
by complement or perforin.75 smoking.175 In RA, inflammation is likely the most impor-
During NET formation, PAD4-mediated histone citrul- tant source of carbamylated proteins. Here, myeloperoxi-
lination is important for chromatin unfolding.171 Activation dase (MPO) released from neutrophils promotes the
of PAD4 during this process is believed to result in citrul- conversion of thiocyanate to cyanate, further enhancing
lination of other molecules, resulting in autoantigen genera- carbamylation.175,176 Although homocitrulline structurally
840 PART 7    DIAGNOSTIC TESTS AND PROCEDURES IN RHEUMATIC DIEASES

resembles citrulline (homocitrulline is one methylene group Because neutrophils produce all components (i.e., PADs,
longer), the finding that anti–carbamylated protein (anti- MPO, and ROS) required to generate the current set of
CarP) antibodies are present in approximately 35% of modified autoantigens in RA (i.e., ACPAs, anti-CarP, and
ACPA− patients and that these antibodies are predictive of anti-MAA), it has been suggested recently that these dis-
a more severe disease course independent of ACPA status tinct antibody systems may indeed represent markers of a
has suggested that these antibodies are not cross-reactive common pathogenic event driven by neutrophils, poten-
between the two PTMs.174 Moreover, anti-CarP antibodies tially placing these cells at the center of autoantigen
appear many years before the diagnosis of RA and predict production in RA pathogenesis.190 Confirming this hypoth-
the development of RA independent of anti-CCP antibod- esis would have important implications for future anti-
ies in patients with arthralgia.177-179 Despite the growing neutrophil therapies in this disease.
interest in anti-CarP antibodies, a major caveat is that their
primary protein targets in RA are still unknown. Indeed, OTHER AUTOANTIBODY SPECIFICITIES IN
carbamylated–fetal calf serum and carbamylated-fibrinogen RHEUMATOID ARTHRITIS RECOGNIZING
are used as surrogate antigens for anti-CarP detection. Fur- NONMODIFIED AUTOANTIGENS
thermore, carbamylated proteins have not yet been described
in the RA joint. Anti-RA33 antibodies were discovered in 1989 as a novel
autoantibody specificity recognizing a nuclear protein with
an apparent molecular mass of 33 kDa.191 These autoanti-
Anti–Malondialdehyde-Acetaldehyde
bodies initially were detected in 35% of patients with RA
Adducts Antibodies
but also were detected in a small number of patients who
Malondialdehyde-acetaldehyde (MAA) adducts are the had other autoimmune or degenerative rheumatic disorders.
most recently described target of anti-PTM antibodies in The antigen was termed RA33, and it was the first descrip-
RA.180 Similar to carbamylation, the generation of MAA tion of a nuclear antigen apparently specific for RA. Later,
protein adducts is nonenzymatic and involves the preferen- the antigen was identified as the A2 protein of the hetero-
tial modification of the primary amine of lysine residues. geneous nuclear ribonucleoprotein complex (hnRNP-
During oxidative stress caused by a variety of stimuli, includ- A2).192 Further characterization of this antibody showed
ing inflammation and exposure to alcohol, reactive oxygen that it was not strictly specific for RA but was also found
species (ROS) trigger peroxidation of membrane lipids, in approximately 20% of patients with SLE and in 40%
resulting in the formation of malondialdehyde (MDA) and to 60% of patients with mixed connective tissue disease
acetaldehyde (AA),181-184 which react with proteins, result- (MCTD).193,194 However, in SLE and MCTD, they usually
ing in the formation of stable MDA-AA protein adducts, occur together with antibodies to U1–small nuclear ribo­
referred to as MAA (Figure 56-3C).185,186 These adducts nucleoproteins (snRNPs) or Smith (Sm) antigen. Therefore
have been implicated in contributing to inflammation and anti-RA33 without concomitant anti–U1 snRNP autoanti-
cellular injury associated with cardiovascular and alcoholic bodies were found to have specificity of 96% for RA.193
liver disease.181-183 Interestingly, RA synovial tissue, but not Additionally, anti-RA33 antibodies in RA, SLE, and
tissue from patients with osteoarthritis, is enriched in MAA- MCTD are distinguished by their recognition of different
modified proteins, and patients with RA have antibodies conformation-dependent epitopes in hnRNP-A2.195 Inter-
targeting this modification when they are detected using estingly, despite their limited specificity, in the absence of
MAA-modified albumin as a surrogate antigen.180 Whereas RF and ACPAs, anti-RA33 antibodies in patients with very
anti-MAA antibodies are strongly associated with anti-CCP early arthritis (<3 months’ duration) are associated with a
and RF, it is interesting that anti-MAA antibodies of the relatively mild nonerosive disease course, potentially iden-
IgG isotype are found in 88% of anti-CCP− patients, sug- tifying patients with a good prognosis who will respond well
gesting that these may be useful biomarkers, particularly in to treatment with disease-modifying anti-rheumatic drugs
persons with ACPA− RA. However, whether this antibody (DMARDs).196 In studies in human samples, stimulation
system is clinically relevant in RA remains to be defined. assays using hnRNP-A2–induced T cell proliferation in
In this regard, it is important to highlight that anti-MAA almost 60% of the patients with RA, but only 20% of the
adduct antibodies are not specific for RA but have also been control subjects, with substantially stronger responses
found in patients with alcohol-induced liver disease, diabe- observed in patients with RA.197 Interestingly, immunohis-
tes, and CVD.187-189 Therefore these antibodies may not tochemical analyses revealed pronounced overexpression of
exclusively be markers of joint damage but may be generated hnRNP-A2 in synovial tissue of patients with RA, placing
as a result of comorbidities associated with RA. Similar to the antigen at sites of disease in RA.
carbamylation, the primary targets of anti-MAA adduct Another autoantigen of potential interest in RA is
antibodies in RA are not yet known. glucose-6-phosphate isomerase (G6PI). Arthritis in the K/
BxN mouse model results from pathogenic immunoglobu-
lins that recognize G6PI,198-200 a glycolytic enzyme residing
Anti-citrullinated Protein Antibodies, Anti–
in the cytoplasm of all cells. Moreover, antibodies directed
Carbamylated Protein, and Anti–
against G6PI can, alone, transfer arthritis to healthy recipi-
Malondialdehyde-Acetaldehyde Antibodies
ents.201 The pathogenic potential of this antibody prompted
in Rheumatoid Arthritis: Insights
its study in human RA. However, although initial studies
into Pathogenesis
reported a high frequency of such antibodies in sera of
The finding that persons with RA have antibodies targeting patients with RA,202 these results have been the subject of
different PTMs is intriguing and unlikely to be accidental. debate.203-205 Overall, anti-G6PI antibodies in patients with
CHAPTER 56    Autoantibodies in Rheumatoid Arthritis 841

RA range between 12% to 29% in different cohorts, with a in ACPAs may influence pathogenicity of these
higher prevalence in patients with active disease. Patients antibodies.216,217
with psoriatic arthritis, undifferentiated arthritis, and
spondylarthropathy also displayed anti-G6PI antibodies at Potential Environmental Factors in
similar frequencies (12% to 25%), and similar titers are Rheumatoid Arthritis Pathogenesis
detected in a proportion (5% to 10%) of control subjects or
patients with Crohn’s disease or sarcoidosis.205 The potential Although abundant citrullination is found in RA synovial
relevance of these antibodies in RA is still uncertain. tissue once disease is established, no studies have addressed
whether abnormal synovial citrullination precedes the
ANTI-CITRULLINATED PROTEIN onset of clinical RA (a requisite to explain the presence of
ANTIBODIES IN RHEUMATOID ARTHRITIS: antibodies against citrullinated proteins long before clinical
INSIGHTS INTO DISEASE MECHANISM disease). Such evidence will be very challenging to obtain.
At this stage, it is therefore important to be aware that the
Although our understanding of the pathogenesis of RA joint is not the only possible site of abnormal citrullination
remains incomplete, current models employ a similar con- that might initiate the RA-specific immune responses to
struct to other autoimmune rheumatic diseases—that is, in citrullinated autoantigens. It is possible that the joint
the setting of a complex genetic predisposition, environ- becomes targeted secondarily after the ACPA immune
mental and stochastic events function to initiate an autoim- response has been initiated at another site, as a consequence
mune process recognizing specific self-antigens, where the of an inflammatory event triggered by a common environ-
immune response itself participates in immune amplifica- mental exposure, such as periodontal infection/inflammation
tion and tissue injury (Figure 56-4). The striking association and/or smoking (Figure 56-4). Although no evidence exists
of RA (and particularly ACPAs) with particular class II that viral proteins are citrullinated in vivo, a subset of
major histocompatibility complex (MHC) alleles under- ACPA+ RA sera has been demonstrated to cross-react with
scores at least one component of this framework. Whether in vitro citrullinated peptides from Epstein-Barr virus
RA autoantibodies are directly pathogenic or whether they EBNA-1 and human papilloma virus (HPV)-47 E2345–362
are markers of inflammation and/or damage remains uncer- proteins.218,219 Defining the kinetics of appearance of these
tain, particularly for RFs. However, ACPAs have several antibodies during disease development, and demonstrating
features that suggest direct involvement in RA pathogenesis directly that these modified proteins are generated during
(Figure 56-3): natural infection, will be important to determine if this
1. The targets of these antibodies (i.e., citrullinated pro- cross-reactivity is relevant to RA pathogenesis.
teins) are abnormally expressed and highly enriched in Tantalizing similarities and epidemiologic associations
synovial tissue and fluid of patients with RA.60,68,133 exist between periodontitis and RA, including the presence
2. The antibody response directed against these citrulli- of bone erosions, association with similar class II MHC
nated antigens precedes RA onset17,104,110,206 and is alleles and smoking, and enrichment of severe periodontal
highly specific for RA and predictive of disease disease in patients with RA.220-224 These connections have
progression.83-87,207 focused attention on the study of oral pathogens that express
3. ACPA positivity is closely linked with the best-known enzymes with PAD activity, specifically Porphyromonas gin-
predisposing genetic risk factors for the development of givalis,225 a Gram-negative bacterium commonly present as
RA: the HLA-DR SE alleles,116,120,208,209 in particular, a biofilm in the gingival crevice and intra-cellularly in oral
HLA-DRB1*04,118,210,211 linking genetic predisposition epithelial cells. P. gingivalis expresses its own citrullinating
and autoantibody production. enzyme named PPAD, which produces ammonia that likely
4. Peptidylcitrulline contained in peptides derived from has negative effects on neutrophil function.226 PPAD is not
aggrecan and vimentin is preferred to arginine in the an ortholog of mammalian PADs, having no sequence
electropositive P4 pocket of the RA-susceptible HLA- homology with mammalian PADs, and showing significant
DRB1*04:01/04, and HLA-II tetramers containing these differences in citrullination activity: (1) PPAD can citrul-
peptides identified circulating citrullinated epitope– linate free L-arginine in addition to protein-bound arginine,
specific CD4+ T cells.212 (2) the enzyme does not require calcium or any other metal
5. Plasma cells producing antibodies against citrullinated ion for activity, and (3) it has preference for citrullination
proteins are present in RA synovial tissue.46 of carboxy-terminal arginines. P. gingivalis also produces
6. About one-half of the anti-CCP+ RA patients have arginine gingipains, endopeptidases that cleave substrates
circulating immune complexes containing citrullinated after arginine residues, a process that generates C-terminal
fibrinogen, and immunohistochemical staining has arginine residues in substrates, which are then citrullinated
shown co-localization of the fibrinogen, immunoglobu- by PPAD.226 Two mechanisms have been proposed by
lin, and complement component C3 in RA pannus which PPAD may be relevant to RA pathogenesis: (1) by
tissue.213 generating citrullinated bacterial and host proteins and
7. Immune complexes containing citrullinated fibrinogen (2) through autocitrullination. The initial finding that
can also activate monocyte-derived macrophages to antibodies to CEP-1 (the immunodominant peptide from
produced TNF via engagement of Fcγ receptors and citrullinated human α-enolase) cross-react with P. gingivalis
Toll-like receptor 4.214,215 Similar observations imply- enolase after citrullination with rabbit PAD opened the
ing overlapping pathogenic properties are likely also possibility that ACPA development might be initially
operative in the case of RFs in RA. Recent evidence driven by citrullinated bacterial products, which subse-
also indicates the that distinct glycosylation patterns quently cross-react with citrullinated endogenous proteins
ASYMPTOMATIC PHASE
A. Initiation Smoking
Viral infection
Environmental factors *
* Bacterial infection
activate the immune system Pg-PAD
*
(e.g., P. gingivalis)
* **
*
P. gingivalis other undefined factors

*
*
PADs + Ca 2+ * Membranolytic cell damage
Citrullination is activated in * (e.g., perforin and MAC), NETs,
immune cells expressing PADs *
Citrullination and others undefined.

PTPN22 (1858 C/T)


Genetic susceptibility YES NO PADI4 polymorphisms
HLA-DR shared epitope alleles
Other undefined genes
Initial ACPA and anti-PAD4
autoantibody response

B. Transition
Augmenting factors drive progression
The ACPA response is expanded over to overt disease phenotype
years and precedes the elevation (e.g., infection, injury, epitope
of inflammatory cytokines Cytokines spreading, isotype switching, etc.)

Asymptomatic Asymptomatic/symptomatic threshold:


Symptomatic When autoantibodies and cytokines
overcome potential compensatory
pathways the disease becomes
clinically evident
CLINICAL PHENOTYPE Autoantibodies bind citrullinated
C. Propagation/damage autoantigens in the joint and activate
inflammatory/damage pathways
(e.g., cellular and humoral immunity, complement, etc.)
*
*

Damaged cells release PADs and


*

PAD4
*
citrullinated autoantigens. * *
* *
PADs continue citrullination *
*
extra-cellularly, which is enhanced by *
activating anti-PAD4 antibodies. * Antigen-presenting cells
*

recognize immune complexes


*

and present citrullinated


*

* peptides to T cells
*
*

Neutrophils and monocytes express


*

PADs, and citrullination is induced


during cell activation, NETs, and
membranolylitic damage

TNF

T cells mediate tissue destruction via the granule pathway, secrete


cytokines, and recruit/activate inflammatory cells
Figure 56-4  A feed-forward model of rheumatoid arthritis (RA) pathogenesis: a pivotal role for protein citrullination. A, During disease initiation,
immune pathways activated against environmental factors (e.g., perforin-induced cytotoxicity, neutrophil extra-cellular traps (NETs), and complement
activation) generate the production of citrullinated proteins in peptidylarginine deiminase (PAD)-expressing cells. Although this process is likely normal
and self-limited in the majority of the cases, some genetically susceptible persons may have the risk of developing autoantibodies against components
involved in the process of citrullination (e.g., PAD4 and citrullinated proteins). This phase is likely asymptomatic because the autoantibodies are in low
titer, they may not be pathogenic, and they target a limited number of citrullinated antigens.78,217 The presence of autoantibodies may persist for
several months to decades, during which a transition phase may occur. In some cases, autoantibody positivity may never progress to RA (i.e., anti-
citrullinated protein antibody [ACPA]+, RA− persons).99,100 B, Depending on the exposure to augmenting factors that promote citrullination (e.g., infec-
tions, smoking, and injury), a transition phase occurs in which the ACPA response is amplified. This phase is characterized by an accumulation of
multiple ACPA specificities, reflecting the process of epitope spreading.78 The expansion of the autoantibody response closely correlates with the
appearance of pre-clinical inflammation that includes the elevation of inflammatory cytokines, such as tumor necrosis factor (TNF), interleukin (IL)-6,
IL-12p70, and interferon γ.78 Moreover, ACPAs acquire a pro-inflammatory Fc glycosylation phenotype prior to the onset of RA.217 Together, the changes
in the autoantibody response and the elevation in cytokines predict the imminent onset of clinical RA.78,217 It is likely that compensatory pathways
might be able to sustain a “healthy state” during the process of transition. However, when autoantibodies and cytokines overcome the threshold of
compensation, the clinical phase is likely initiated. C, Once initiated, the immune response drives a feed-forward loop of target tissue destruction and
disease propagation, resulting in the clinical phenotype. Although the citrullinating events that initiate the ACPA response may occur outside the joint,
during disease propagation, citrullinated autoantigens are generated in the synovium by inflammatory cell PADs, leading to amplification of the
immune response within the target tissue. Citrullinated antigens are indicated (*). HLA-DR, Human leukocyte antigen–DR region; MAC, membrane
attack complex.
CHAPTER 56    Autoantibodies in Rheumatoid Arthritis 843

in the microenvironment of the inflamed joint.227 Subse- 7. Shmerling RH, Delbanco TL: How useful is the rheumatoid factor?
quently, it was found that arginine gingipain and PPAD, An analysis of sensitivity, specificity, and predictive value. Arch Intern
Med 152:2417–2420, 1992.
respectively, cleave and citrullinate human fibrinogen and 8. Dorner T, Egerer K, Feist E, et al: Rheumatoid factor revisited. Curr
α-enolase in vitro, extending this hypothesis that host pro- Opin Rheumatol 16:246–253, 2004.
teins citrullinated by the bacterial enzyme may also play a 11. Randen I, et al: Rheumatoid factor V genes from patients with rheu-
role in initiating the ACPA response in RA.228 Although matoid arthritis are diverse and show evidence of an antigen-driven
response. Immunol Rev 128:49–71, 1992.
this model is intriguing, the citrullination of bacterial 14. Nishimura K, et al: Meta-analysis: diagnostic accuracy of anti-cyclic
enolase by PPAD or by a human host PAD during bacterial citrullinated peptide antibody and rheumatoid factor for rheumatoid
lysis of phagocytic cells has not been demonstrated. In addi- arthritis. Ann Intern Med 146:797–808, 2007.
tion, it is unclear whether the C-terminal citrullinated 17. Nielen MM, et al: Specific autoantibodies precede the symptoms of
human fibrinogen and α-enolase fragments generated by rheumatoid arthritis: a study of serial measurements in blood donors.
Arthritis Rheum 50:380–386, 2004.
gingipains and PPAD are indeed recognized by ACPAs 18. Halldorsdottir HD, Jonsson T, Thorsteinsson J, et al: A prospective
in RA. An alternative mechanism that has been proposed study on the incidence of rheumatoid arthritis among people with
for the involvement of PPAD in RA pathogenesis is that persistent increase of rheumatoid factor. Ann Rheum Dis 59:149–151,
autocitrullinated PPAD may act as the inciting citrullinated 2000.
19. Jansen LM, van der Horst-Bruinsma IE, van Schaardenburg D, et al:
antigen that may trigger the ACPA response in RA.229 Predictors of radiographic joint damage in patients with early rheu-
Although this theory is provocative, it remains controver- matoid arthritis. Ann Rheum Dis 60:924–927, 2001.
sial because PPAD autocitrullination appears to be an arti- 21. Nienhuis RL, Mandema E: A new serum factor in patients with
fact generated when the protein is artificially expressed in rheumatoid arthritis; the antiperinuclear factor. Ann Rheum Dis
Escherichia coli.230,231 23:302–305, 1964.
22. Sondag-Tschroots IR, Aaij C, Smit JW, et al: The antiperinuclear
Another environmental factor with potential relevance factor. 1. The diagnostic significance of the antiperinuclear factor for
in initiating production of ACPAs is cigarette smoking. rheumatoid arthritis. Ann Rheum Dis 38:248–251, 1979.
Interestingly, recent evidence has demonstrated (at least in 23. Hoet RM, Boerbooms AM, Arends M, et al: Antiperinuclear factor,
some populations) a strong interaction between smoking a marker autoantibody for rheumatoid arthritis: colocalisation of the
perinuclear factor and profilaggrin. Ann Rheum Dis 50:611–618,
and SE alleles in the development of ACPA+ RA.119,232-234 1991.
However, whereas primarily HLA-DRB1*04 alleles have 31. Aggarwal R, Liao K, Nair R, et al: Anti-citrullinated peptide anti-
been associated with ACPA production,118,210,211 in smokers, body assays and their role in the diagnosis of rheumatoid arthritis.
HLA-DRB1*0101, *0102, and *1001 alleles showed the Arthritis Rheum 61:1472–1483, 2009.
closest association with positive anti-CCP.119 The observa- 32. Young BJ, Mallya RK, Leslie RD, et al: Anti-keratin antibodies in
rheumatoid arthritis. Br Med J 2:97–99, 1979.
tion that cigarette smoking induces increased expression of 37. Paimela L, Gripenberg M, Kurki P, et al: Antikeratin antibodies:
PAD2 and protein citrullination in lungs,51 that anti-CCP diagnostic and prognostic markers for early rheumatoid arthritis. Ann
antibodies of the IgA class are found in about one-third of Rheum Dis 51:743–746, 1992.
patients with recent-onset RA,235 and that anti-CCP anti- 38. Simon M, et al: The cytokeratin filament-aggregating protein filla-
grin is the target of the co-called “antikeratin antibodies,” autoanti-
bodies can be found in patients with ILD who do not have bodies specific for rheumatoid arthritis. J Clin Invest 92:1387–1393,
RA99,100 has suggested that the lung or other mucosal sur- 1993.
faces may play a role in generating or sustaining autoanti- 39. Sebbag M, et al: The antiperinuclear factor and the so-called antik-
bodies to citrullinated proteins. These gene-environment eratin antibodies are the same rheumatoid arthritis-specific autoan-
interactions, playing out in microenvironments outside the tibodies. J Clin Invest 95:2672–2679, 1995.
43. Vossenaar ER, Zendman AJ, van Venrooij WJ, et al: PAD, a growing
joint, add important dimensions to the framework within family of citrullinating enzymes: genes, features and involvement in
which RA pathogenesis must be investigated and under- disease. Bioessays 25:1106–1118, 2003.
stood. Although it is possible that none of the detailed 44. Schellekens GA, de Jong BA, van den Hoogen FH, et al: Citrulline
mechanisms currently defined represent the unifying is an essential constituent of antigenic determinants recognized by
rheumatoid arthritis-specific autoantibodies. J Clin Invest 101:273–
mechanism of pathogenesis in RA, the broader view of 281, 1998.
potential immunizing environments and the search for a 45. Baeten D, et al: Specific presence of intracellular citrullinated pro-
frequent worldwide environmental exposure is of great teins in rheumatoid arthritis synovium: relevance to antifilaggrin
importance. autoantibodies. Arthritis Rheum 44:2255–2262, 2001.
46. Masson-Bessiere C, et al: In the rheumatoid pannus, anti-filaggrin
autoantibodies are produced by local plasma cells and constitute a
higher proportion of IgG than in synovial fluid and serum. Clin Exp
The full references for this chapter can be found on Immunol 119:544–552, 2000.
ExpertConsult.com. 49. Chapuy-Regaud S, et al: Fibrin deimination in synovial tissue is not
specific for rheumatoid arthritis but commonly occurs during syno-
vitides. J Immunol 174:5057–5064, 2005.
50. Makrygiannakis D, et al: Citrullination is an inflammation-dependent
SELECTED REFERENCES process. Ann Rheum Dis 65:1219–1222, 2006.
1. Waaler E: On the occurrence of a factor in human serum activating 54. Schellekens GA, et al: The diagnostic properties of rheumatoid
the specific agglutintion of sheep blood corpuscles. 1939. APMIS arthritis antibodies recognizing a cyclic citrullinated peptide. Arthritis
115:422–438, 2007. Rheum 43:155–163, 2000.
2. Rose HM, Ragan C, et al: Differential agglutination of normal and 55. van Venrooij WJ, Zendman AJ: Anti-CCP2 antibodies: an overview
sensitized sheep erythrocytes by sera of patients with rheumatoid and perspective of the diagnostic abilities of this serological marker
arthritis. Proc Soc Exp Biol Med 68:1–6, 1948. for early rheumatoid arthritis. Clin Rev Allergy Immunol 34:36–39,
3. Henney CS, Stanworth DR: Reaction of rheumatoid factor with the 2008.
isolated polypeptide chains of human 7S gamma-globulin. Nature 56. van Gaalen FA, Visser H, Huizinga TW: A comparison of the diag-
201:511–512, 1964. nostic accuracy and prognostic value of the first and second anti-
6. Shmerling RH, Delbanco TL: The rheumatoid factor: an analysis of cyclic citrullinated peptides (CCP1 and CCP2) autoantibody tests
clinical utility. Am J Med 91:528–534, 1991. for rheumatoid arthritis. Ann Rheum Dis 64:1510–1512, 2005.
844 PART 7    DIAGNOSTIC TESTS AND PROCEDURES IN RHEUMATIC DIEASES

57. Lutteri L, Malaise M, Chapelle JP: Comparison of second- and third- 89. Kelly CA, et al: Rheumatoid arthritis-related interstitial lung disease:
generation anti-cyclic citrullinated peptide antibodies assays for associations, prognostic factors and physiological and radiological
detecting rheumatoid arthritis. Clin Chim Acta 386:76–81, 2007. characteristics—a large multicentre UK study. Rheumatology (Oxford)
58. dos Anjos LM, et al: A comparative study of IgG second- and third- 53:1676–1682, 2014.
generation anti-cyclic citrullinated peptide (CCP) ELISAs and their 90. Giles JT, et al: Association of fine specificity and repertoire expan-
combination with IgA third-generation CCP ELISA for the diagnosis sion of anticitrullinated peptide antibodies with rheumatoid arthritis
of rheumatoid arthritis. Clin Rheumatol 28:153–158, 2009. associated interstitial lung disease. Ann Rheum Dis 73:1487–1494,
59. Masson-Bessière C, et al: The major synovial targets of the rheuma- 2014.
toid arthritis-specific antifilaggrin autoantibodies are deiminated 91. Aubart F, et al: High levels of anti-cyclic citrullinated peptide auto-
forms of the alpha- and beta-chains of fibrin. J Immunol 166:4177– antibodies are associated with co-occurrence of pulmonary diseases
4184, 2001. with rheumatoid arthritis. J Rheumatol 38:979–982, 2011.
62. Vossenaar ER, et al: Rheumatoid arthritis specific anti-Sa antibodies 92. Zhu J, Zhou Y, Chen X, et al: A metaanalysis of the increased risk of
target citrullinated vimentin. Arthritis Res Ther 6:R142–R150, 2004. rheumatoid arthritis-related pulmonary disease as a result of serum
63. van Beers JJ, et al: Anti-citrullinated fibronectin antibodies in rheu- anticitrullinated protein antibody positivity. J Rheumatol 41:1282–
matoid arthritis are associated with human leukocyte antigen-DRB1 1289, 2014.
shared epitope alleles. Arthritis Res Ther 14:R35, 2012. 94. Marasovic-Krstulovic D, Martinovic-Kaliterna D, Fabijanic D, et al:
64. Darrah E, Rosen A, Giles JT, et al: Peptidylarginine deiminase 2, 3 Are the anti-cyclic citrullinated peptide antibodies independent pre-
and 4 have distinct specificities against cellular substrates: novel dictors of myocardial involvement in patients with active rheumatoid
insights into autoantigen selection in rheumatoid arthritis. Ann arthritis? Rheumatology (Oxford) 50:1505–1512, 2011.
Rheum Dis 71:92–98, 2012. 97. Bongartz T, et al: Citrullination in extra-articular manifestations of
67. Kinloch A, et al: Identification of citrullinated alpha-enolase as a rheumatoid arthritis. Rheumatology (Oxford) 46:70–75, 2007.
candidate autoantigen in rheumatoid arthritis. Arthritis Res Ther 99. Gizinski AM, et al: Rheumatoid arthritis (RA)-specific autoantibod-
7:R1421–R1429, 2005. ies in patients with interstitial lung disease and absence of clinically
73. van Beers JJ, et al: The rheumatoid arthritis synovial fluid citrulli- apparent articular RA. Clin Rheumatol 28:611–613, 2009.
nome reveals novel citrullinated epitopes in apolipoprotein E, 101. Visser K, et al: Pretreatment serum levels of anti-cyclic citrullinated
myeloid nuclear differentiation antigen, and beta-actin. Arthritis peptide antibodies are associated with the response to methotrexate
Rheum 65:69–80, 2013. in recent-onset arthritis. Ann Rheum Dis 67:1194–1195, 2008.
74. Saulot V, et al: Presence of autoantibodies to the glycolytic enzyme 102. Farragher TM, et al: Benefit of early treatment in inflammatory poly-
alpha-enolase in sera from patients with early rheumatoid arthritis. arthritis patients with anti-cyclic citrullinated peptide antibodies
Arthritis Rheum 46:1196–1201, 2002. versus those without antibodies. Arthritis Care Res (Hoboken) 62:
75. Romero V, et al: Immune-mediated pore-forming pathways induce 664–675, 2010.
cellular hypercitrullination and generate citrullinated autoantigens 103. Braun-Moscovici Y, et al: Anti-cyclic citrullinated protein antibodies
in rheumatoid arthritis. Sci Transl Med 5:209ra150, 2013. as a predictor of response to anti-tumor necrosis factor-alpha therapy
76. Tutturen AE, Fleckenstein B, de Souza GA: Assessing the citrulli- in patients with rheumatoid arthritis. J Rheumatol 33:497–500, 2006.
nome in rheumatoid arthritis synovial fluid with and without enrich- 107. Kolfenbach JR, et al: Autoimmunity to peptidyl arginine deiminase
ment of citrullinated peptides. J Proteome Res 13:2867–2873, 2014. type 4 precedes clinical onset of rheumatoid arthritis. Arthritis Rheum
77. Bang H, et al: Mutation and citrullination modifies vimentin to a 62:2633–2639, 2010.
novel autoantigen for rheumatoid arthritis. Arthritis Rheum 56:2503– 109. Kokkonen H, et al: Antibodies of IgG, IgA and IgM isotypes against
2511, 2007. cyclic citrullinated peptide precede the development of rheumatoid
78. Sokolove J, et al: Autoantibody epitope spreading in the pre-clinical arthritis. Arthritis Res Ther 13:R13, 2011.
phase predicts progression to rheumatoid arthritis. PLoS One 110. Rantapaa-Dahlqvist S, et al: Antibodies against cyclic citrullinated
7:e35296, 2012. peptide and IgA rheumatoid factor predict the development of rheu-
79. Wagner CA, et al: Identification of anticitrullinated protein anti- matoid arthritis. Arthritis Rheum 48:2741–2749, 2003.
body reactivities in a subset of anti-CCP-negative rheumatoid arthri- 111. Kolfenbach JR, et al: A prospective approach to investigating the
tis: association with cigarette smoking and HLA-DRB1 “shared natural history of preclinical rheumatoid arthritis (RA) using first-
epitope” alleles. Ann Rheum Dis 74:579–586, 2015. degree relatives of probands with RA. Arthritis Rheum 61:1735–1742,
80. van Gaalen FA, et al: Autoantibodies to cyclic citrullinated peptides 2009.
predict progression to rheumatoid arthritis in patients with undif- 112. Berglin E, et al: Radiological outcome in rheumatoid arthritis is pre-
ferentiated arthritis: a prospective cohort study. Arthritis Rheum dicted by presence of antibodies against cyclic citrullinated peptide
50:709–715, 2004. before and at disease onset, and by IgA-RF at disease onset. Ann
81. Jansen LM, et al: The predictive value of anti-cyclic citrullinated Rheum Dis 65:453–458, 2006.
peptide antibodies in early arthritis. J Rheumatol 30:1691–1695, 2003. 113. Majka DS, et al: Duration of preclinical rheumatoid arthritis-related
82. Nell VP, et al: Autoantibody profiling as early diagnostic and prog- autoantibody positivity increases in subjects with older age at time
nostic tool for rheumatoid arthritis. Ann Rheum Dis 64:1731–1736, of disease diagnosis. Ann Rheum Dis 67:801–807, 2008.
2005. 114. Gregersen PK, Silver J, Winchester RJ: The shared epitope hypoth-
83. Vencovsky J, et al: Autoantibodies can be prognostic markers of an esis. An approach to understanding the molecular genetics of suscep-
erosive disease in early rheumatoid arthritis. Ann Rheum Dis 62:427– tibility to rheumatoid arthritis. Arthritis Rheum 30:1205–1213, 1987.
430, 2003. 115. van der Helm-van Mil AH, Huizinga TW: Advances in the genetics
84. Mewar D, et al: Independent associations of anti-cyclic citrullinated of rheumatoid arthritis point to subclassification into distinct disease
peptide antibodies and rheumatoid factor with radiographic severity subsets. Arthritis Res Ther 10:205, 2008.
of rheumatoid arthritis. Arthritis Res Ther 8:R128, 2006. 116. Huizinga TW, et al: Refining the complex rheumatoid arthritis phe-
85. Syversen SW, et al: High anti-cyclic citrullinated peptide levels and notype based on specificity of the HLA-DRB1 shared epitope for
an algorithm of four variables predict radiographic progression in antibodies to citrullinated proteins. Arthritis Rheum 52:3433–3438,
patients with rheumatoid arthritis: results from a 10-year longitudinal 2005.
study. Ann Rheum Dis 67:212–217, 2008. 117. Irigoyen P, et al: Regulation of anti-cyclic citrullinated peptide anti-
86. De Rycke L, et al: Rheumatoid factor and anticitrullinated protein bodies in rheumatoid arthritis: contrasting effects of HLA-DR3 and
antibodies in rheumatoid arthritis: diagnostic value, associations with the shared epitope alleles. Arthritis Rheum 52:3813–3818, 2005.
radiological progression rate, and extra-articular manifestations. Ann 118. Snir O, et al: Multiple antibody reactivities to citrullinated antigens
Rheum Dis 63:1587–1593, 2004. in sera from patients with rheumatoid arthritis: association with
87. Turesson C, et al: Rheumatoid factor and antibodies to cyclic citrul- HLA-DRB1 alleles. Ann Rheum Dis 68:736–743, 2009.
linated peptides are associated with severe extra-articular manifesta- 119. van der Helm-van Mil AH, et al: The HLA-DRB1 shared epitope
tions in rheumatoid arthritis. Ann Rheum Dis 66:59–64, 2007. alleles differ in the interaction with smoking and predisposition to
88. Quinn MA, et al: Anti-CCP antibodies measured at disease onset antibodies to cyclic citrullinated peptide. Arthritis Rheum 56:425–
help identify seronegative rheumatoid arthritis and predict radiologi- 432, 2007.
cal and functional outcome. Rheumatology (Oxford) 45:478–480, 120. Johansson M, Arlestig L, Hallmans G, et al: PTPN22 polymorphism
2006. and anti-cyclic citrullinated peptide antibodies in combination
CHAPTER 56    Autoantibodies in Rheumatoid Arthritis 845

strongly predicts future onset of rheumatoid arthritis and has a speci- autoantigen modifications in rheumatoid arthritis. Arthritis Rheuma-
ficity of 100% for the disease. Arthritis Res Ther 8:R19, 2006. tol. 67:604–608, 2015.
121. Stanford SM, Mustelin TM, Bottini N: Lymphoid tyrosine phospha- 191. Hassfeld W, et al: Demonstration of a new antinuclear antibody
tase and autoimmunity: human genetics rediscovers tyrosine phos- (anti-RA33) that is highly specific for rheumatoid arthritis. Arthritis
phatases. Semin Immunopathol 32:127–136, 2010. Rheum 32:1515–1520, 1989.
122. Shirai H, Blundell TL, Mizuguchi K: A novel superfamily of enzymes 196. Nell-Duxneuner V, et al: Autoantibody profiling in patients with
that catalyze the modification of guanidino groups. Trends Biochem very early rheumatoid arthritis: a follow-up study. Ann Rheum Dis
Sci 26:465–468, 2001. 69:169–174, 2010.
128. Arita K, et al: Structural basis for Ca(2+)-induced activation of 198. Kouskoff V, et al: Organ-specific disease provoked by systemic auto-
human PAD4. Nat Struct Mol Biol 11:777–783, 2004. immunity. Cell 87:811–822, 1996.
132. Foulquier C, et al: Peptidyl arginine deiminase type 2 (PAD-2) and 203. Kassahn D, Kolb C, Solomon S, et al: Few human autoimmune sera
PAD-4 but not PAD-1, PAD-3, and PAD-6 are expressed in rheuma- detect GPI. Nat Immunol 3:411–412, 2002.
toid arthritis synovium in close association with tissue inflammation. 205. Matsumoto I, et al: Low prevalence of antibodies to glucose-6-
Arthritis Rheum 56:3541–3553, 2007. phosphate isomerase in patients with rheumatoid arthritis and a
133. Chang X, et al: Localization of peptidylarginine deiminase 4 (PADI4) spectrum of other chronic autoimmune disorders. Arthritis Rheum
and citrullinated protein in synovial tissue of rheumatoid arthritis. 48:944–954, 2003.
Rheumatology (Oxford) 44:40–50, 2005. 207. Zendman AJ, Vossenaar ER, van Venrooij WJ: Autoantibodies to
136. Darrah E, et al: Erosive rheumatoid arthritis is associated with anti- citrullinated (poly)peptides: a key diagnostic and prognostic marker
bodies that activate PAD4 by increasing calcium sensitivity. Sci Transl for rheumatoid arthritis. Autoimmunity 37:295–299, 2004.
Med 5:186ra65, 2013. 208. Kokkonen H, Johansson M, Innala L, et al: The PTPN22 1858C/T
141. Andrade F, et al: Autocitrullination of human peptidyl arginine polymorphism is associated with anti-cyclic citrullinated peptide
deiminase type 4 regulates protein citrullination during cell activa- antibody-positive early rheumatoid arthritis in northern Sweden.
tion. Arthritis Rheum 62:1630–1640, 2010. Arthritis Res Ther 9:R56, 2007.
142. Mechin MC, et al: Deimination is regulated at multiple levels includ- 209. van der Helm-van Mil AH, et al: The HLA-DRB1 shared epitope
ing auto-deimination of peptidylarginine deiminases. Cell Mol Life Sci alleles are primarily a risk factor for anti-cyclic citrullinated peptide
67:1491–1503, 2010. antibodies and are not an independent risk factor for development
147. Loos T, et al: Citrullination of CXCL10 and CXCL11 by peptidylar- of rheumatoid arthritis. Arthritis Rheum 54:1117–1121, 2006.
ginine deiminase: a naturally occurring posttranslational modifica- 210. van der Helm-van Mil AH, Wesoly JZ, Huizinga TW: Understanding
tion of chemokines and new dimension of immunoregulation. Blood the genetic contribution to rheumatoid arthritis. Curr Opin Rheuma-
112:2648–2656, 2008. tol 17:299–304, 2005.
155. Li P, et al: PAD4 is essential for antibacterial innate immunity medi- 212. Scally SW, et al: A molecular basis for the association of the HLA-
ated by neutrophil extracellular traps. J Exp Med 207:1853–1862, DRB1 locus, citrullination, and rheumatoid arthritis. J Exp Med
2010. 210:2569–2582, 2013.
157. Suzuki A, et al: Functional haplotypes of PADI4, encoding citrulli- 213. Zhao X, et al: Circulating immune complexes contain citrullinated
nating enzyme peptidylarginine deiminase 4, are associated with fibrinogen in rheumatoid arthritis. Arthritis Res Ther 10:R94, 2008.
rheumatoid arthritis. Nat Genet 34:395–402, 2003. 214. Clavel C, et al: Induction of macrophage secretion of tumor necrosis
163. Harris ML, et al: Association of autoimmunity to peptidyl arginine factor alpha through Fcgamma receptor IIa engagement by rheuma-
deiminase type 4 with genotype and disease severity in rheumatoid toid arthritis-specific autoantibodies to citrullinated proteins com-
arthritis. Arthritis Rheum 58:1958–1967, 2008. plexed with fibrinogen. Arthritis Rheum 58:678–688, 2008.
164. Bang SY, et al: Peptidyl arginine deiminase type IV (PADI4) haplo- 215. Sokolove J, Zhao X, Chandra PE, et al: Immune complexes contain-
types interact with shared epitope regardless of anti-cyclic citrulli- ing citrullinated fibrinogen costimulate macrophages via Toll-like
nated peptide antibody or erosive joint status in rheumatoid arthritis: receptor 4 and Fc gamma receptor. Arthritis Rheum 63:53–62, 2011.
a case control study. Arthritis Res Ther 12:R115, 2010. 217. Rombouts Y, et al: Anti-citrullinated protein antibodies acquire a
165. Suzuki T, et al: PADI4 and HLA-DRB1 are genetic risks for radio- pro-inflammatory Fc glycosylation phenotype prior to the onset of
graphic progression in RA patients, independent of ACPA status: rheumatoid arthritis. Ann Rheum Dis 74:234–241, 2015.
results from the IORRA cohort study. PLoS One 8:e61045, 2013. 221. Kasser UR, et al: Risk for periodontal disease in patients with
166. Halvorsen EH, et al: Serum IgG antibodies to peptidylarginine deim- longstanding rheumatoid arthritis. Arthritis Rheum 40:2248–2251,
inase 4 in rheumatoid arthritis and associations with disease severity. 1997.
Ann Rheum Dis 67:414–417, 2008. 223. de Pablo P, Chapple IL, Buckley CD, et al: Periodontitis in systemic
167. Halvorsen EH, et al: Serum IgG antibodies to peptidylarginine deim- rheumatic diseases. Nat Rev Rheumatol 5:218–224, 2009.
inase 4 predict radiographic progression in patients with rheumatoid 224. Lundberg K, Wegner N, Yucel-Lindberg T, et al: Periodontitis in
arthritis treated with tumour necrosis factor-alpha blocking agents. RA—the citrullinated enolase connection. Nat Rev Rheumatol
Ann Rheum Dis 68:249–252, 2009. 6:727–730, 2010.
168. Giles JT, et al: Association of cross-reactive antibodies targeting 227. Lundberg K, et al: Antibodies to citrullinated alpha-enolase peptide
peptidyl-arginine deiminase 3 and 4 with rheumatoid arthritis- 1 are specific for rheumatoid arthritis and cross-react with bacterial
associated interstitial lung disease. PLoS One 9:e98794, 2014. enolase. Arthritis Rheum 58:3009–3019, 2008.
170. Khandpur R, et al: NETs are a source of citrullinated autoantigens 228. Wegner N, et al: Peptidylarginine deiminase from Porphyromonas
and stimulate inflammatory responses in rheumatoid arthritis. Sci gingivalis citrullinates human fibrinogen and alpha-enolase: implica-
Transl Med 5:178ra40, 2013. tions for autoimmunity in rheumatoid arthritis. Arthritis Rheum 62:
173. Damgaard D, Senolt L, Nielsen MF, et al: Demonstration of extracel- 2662–2672, 2010.
lular peptidylarginine deiminase (PAD) activity in synovial fluid of 230. Konig MF, Paracha AS, Moni M, et al: Defining the role of Porphy-
patients with rheumatoid arthritis using a novel assay for citrullina- romonas gingivalis peptidylarginine deiminase (PPAD) in rheumatoid
tion of fibrinogen. Arthritis Res Ther 16:498, 2014. arthritis through the study of PPAD biology. Ann Rheum Dis pii:ann
174. Shi J, et al: Autoantibodies recognizing carbamylated proteins are rheumdis-2014-205385, May 26, 2014. 74:2054–61, 2015.
present in sera of patients with rheumatoid arthritis and predict joint 232. Klareskog L, et al: A new model for an etiology of rheumatoid
damage. Proc Natl Acad Sci U S A 108:17372–17377, 2011. arthritis: smoking may trigger HLA-DR (shared epitope)-restricted
177. Shi J, et al: Anti-carbamylated protein (anti-CarP) antibodies immune reactions to autoantigens modified by citrullination. Arthritis
precede the onset of rheumatoid arthritis. Ann Rheum Dis 73:780– Rheum 54:38–46, 2006.
783, 2014. 233. Linn-Rasker SP, et al: Smoking is a risk factor for anti-CCP antibod-
180. Thiele GM, et al: Malondialdehyde-acetaldehyde adducts and anti- ies only in rheumatoid arthritis patients who carry HLA-DRB1
malondialdehyde-acetaldehyde antibodies in rheumatoid arthritis. shared epitope alleles. Ann Rheum Dis 65:366–371, 2006.
Arthritis Rheumatol 67:645–655, 2015. 234. Klareskog L, Malmstrom V, Lundberg K, et al: Smoking, citrullina-
190. Darrah E, Andrade F: Editorial: citrullination, and carbamylation, tion and genetic variability in the immunopathogenesis of rheuma-
and malondialdehyde-acetaldehyde! Oh my! Entering the forest of toid arthritis. Semin Immunol 23:92–98, 2011.
CHAPTER 56    Autoantibodies in Rheumatoid Arthritis 845.e1

27. Manera C, Franceschini F, Cretti L, et al: Clinical heterogeneity of


REFERENCES rheumatoid arthritis and the antiperinuclear factor. J Rheumatol 21:
1. Waaler E: On the occurrence of a factor in human serum activating 2021–2025, 1994.
the specific agglutintion of sheep blood corpuscles. 1939. APMIS 28. Youinou P, et al: The antiperinuclear factor. I. Clinical and serologic
115:422–438, 2007. associations. Clin Exp Rheumatol 8:259–264, 1990.
2. Rose HM, Ragan C, et al: Differential agglutination of normal and 29. Youinou P, et al: The antiperinuclear factor. II. Variability of the
sensitized sheep erythrocytes by sera of patients with rheumatoid perinuclear antigen. Clin Exp Rheumatol 8:265–269, 1990.
arthritis. Proc Soc Exp Biol Med 68:1–6, 1948. 30. Youinou P, Le GP: The reliability of the antiperinuclear factor test
3. Henney CS, Stanworth DR: Reaction of rheumatoid factor with the despite the inconstancy of the targeted antigens. J Rheumatol 21:
isolated polypeptide chains of human 7S gamma-globulin. Nature 1990–1991, 1994.
201:511–512, 1964. 31. Aggarwal R, Liao K, Nair R, et al: Anti-citrullinated peptide anti-
4. Plotz CM, Singer JM: The latex fixation test. I. Application to the body assays and their role in the diagnosis of rheumatoid arthritis.
serologic diagnosis of rheumatoid arthritis. Am J Med 21:888–892, Arthritis Rheum 61:1472–1483, 2009.
1956. 32. Young BJ, Mallya RK, Leslie RD, et al: Anti-keratin antibodies in
5. Plotz CM, Singer JM: The latex fixation test. II. Results in rheuma- rheumatoid arthritis. Br Med J 2:97–99, 1979.
toid arthritis. Am J Med 21:893–896, 1956. 33. Scott DL, Delamere JP, Jones LJ, et al: Significance of laminar anti-
6. Shmerling RH, Delbanco TL: The rheumatoid factor: an analysis of keratin antibodies to rat oesophagus in rheumatoid arthritis. Ann
clinical utility. Am J Med 91:528–534, 1991. Rheum Dis 40:267–271, 1981.
7. Shmerling RH, Delbanco TL: How useful is the rheumatoid factor? 34. Miossec P, Youinou P, Le GP, et al: Clinical relevance of antikera-
An analysis of sensitivity, specificity, and predictive value. Arch Intern tin antibodies in rheumatoid arthritis. Clin Rheumatol 1:185–189,
Med 152:2417–2420, 1992. 1982.
8. Dorner T, Egerer K, Feist E, et al: Rheumatoid factor revisited. Curr 35. Ordeig J, Guardia J: Diagnostic value of antikeratin antibodies in
Opin Rheumatol 16:246–253, 2004. rheumatoid arthritis. J Rheumatol 11:602–604, 1984.
9. Prentice AG, Hickling P, Wiseman IC, et al: Prospective comparison 36. Vincent C, et al: High diagnostic value in rheumatoid arthritis of
of laser nephelometry with standard agglutination techniques for antibodies to the stratum corneum of rat oesophagus epithelium,
detection of rheumatoid factor. J Clin Pathol 40:216–220, 1987. so-called “antikeratin antibodies.” Ann Rheum Dis 48:712–722, 1989.
10. Anuradha V, Chopra A: In the era of nephelometry, latex agglutina- 37. Paimela L, Gripenberg M, Kurki P, et al: Antikeratin antibodies:
tion is still good enough to detect rheumatoid factor. J Rheumatol diagnostic and prognostic markers for early rheumatoid arthritis. Ann
32:2343–2344, 2005. Rheum Dis 51:743–746, 1992.
11. Randen I, et al: Rheumatoid factor V genes from patients with rheu- 38. Simon M, et al: The cytokeratin filament-aggregating protein filla-
matoid arthritis are diverse and show evidence of an antigen-driven grin is the target of the co-called “antikeratin antibodies,” autoanti-
response. Immunol Rev 128:49–71, 1992. bodies specific for rheumatoid arthritis. J Clin Invest 92:1387–1393,
12. Randen I, et al: Synovial IgG rheumatoid factors show evidence of 1993.
an antigen-driven immune response and a shift in the V gene reper- 39. Sebbag M, et al: The antiperinuclear factor and the so-called antik-
toire compared to IgM rheumatoid factors. Eur J Immunol 23:1220– eratin antibodies are the same rheumatoid arthritis-specific autoan-
1225, 1993. tibodies. J Clin Invest 95:2672–2679, 1995.
13. Jonsson T, et al: Combined elevation of IgM and IgA rheumatoid 40. Markova NG, et al: Profilaggrin is a major epidermal calcium-binding
factor has high diagnostic specificity for rheumatoid arthritis. Rheu- protein. Mol Cell Biol 13:613–625, 1993.
matol Int 18:119–122, 1998. 41. Senshu T, et al: Detection of deiminated proteins in rat skin: probing
14. Nishimura K, et al: Meta-analysis: diagnostic accuracy of anti-cyclic with a monospecific antibody after modification of citrulline residues.
citrullinated peptide antibody and rheumatoid factor for rheumatoid J Invest Dermatol 105:163–169, 1995.
arthritis. Ann Intern Med 146:797–808, 2007. 42. Senshu T, Kan S, Ogawa H, et al: Preferential deimination of keratin
15. Aho K, et al: When does rheumatoid disease start? Arthritis Rheum K1 and filaggrin during the terminal differentiation of human epider-
28:485–489, 1985. mis. Biochem Biophys Res Commun 225:712–719, 1996.
16. Aho K, Heliovaara M, Maatela J, et al: Rheumatoid factors antedat- 43. Vossenaar ER, Zendman AJ, van Venrooij WJ, et al: PAD, a growing
ing clinical rheumatoid arthritis. J Rheumatol 18:1282–1284, 1991. family of citrullinating enzymes: genes, features and involvement in
17. Nielen MM, et al: Specific autoantibodies precede the symptoms of disease. Bioessays 25:1106–1118, 2003.
rheumatoid arthritis: a study of serial measurements in blood donors. 44. Schellekens GA, de Jong BA, van den Hoogen FH, et al: Citrulline
Arthritis Rheum 50:380–386, 2004. is an essential constituent of antigenic determinants recognized by
18. Halldorsdottir HD, Jonsson T, Thorsteinsson J, et al: A prospective rheumatoid arthritis-specific autoantibodies. J Clin Invest 101:273–
study on the incidence of rheumatoid arthritis among people with 281, 1998.
persistent increase of rheumatoid factor. Ann Rheum Dis 59:149–151, 45. Baeten D, et al: Specific presence of intracellular citrullinated pro-
2000. teins in rheumatoid arthritis synovium: relevance to antifilaggrin
19. Jansen LM, van der Horst-Bruinsma IE, van Schaardenburg D, et al: autoantibodies. Arthritis Rheum 44:2255–2262, 2001.
Predictors of radiographic joint damage in patients with early rheu- 46. Masson-Bessière C, et al: In the rheumatoid pannus, anti-filaggrin
matoid arthritis. Ann Rheum Dis 60:924–927, 2001. autoantibodies are produced by local plasma cells and constitute a
20. Panayi GS: B cells: a fundamental role in the pathogenesis of rheu- higher proportion of IgG than in synovial fluid and serum. Clin Exp
matoid arthritis? Rheumatology (Oxford) 44(Suppl 2):ii3–ii7, 2005. Immunol 119:544–552, 2000.
21. Nienhuis RL, Mandema E: A new serum factor in patients with 47. Wood DD, et al: Myelin localization of peptidylarginine deiminases
rheumatoid arthritis; the antiperinuclear factor. Ann Rheum Dis 2 and 4: comparison of PAD2 and PAD4 activities. Lab Invest 88:354–
23:302–305, 1964. 364, 2008.
22. Sondag-Tschroots IR, Aaij C, Smit JW, et al: The antiperinuclear 48. Chavanas S, et al: Peptidylarginine deiminases and deimination in
factor. 1. The diagnostic significance of the antiperinuclear factor for biology and pathology: relevance to skin homeostasis. J Dermatol Sci
rheumatoid arthritis. Ann Rheum Dis 38:248–251, 1979. 44:63–72, 2006.
23. Hoet RM, Boerbooms AM, Arends M, et al: Antiperinuclear factor, 49. Chapuy-Regaud S, et al: Fibrin deimination in synovial tissue is not
a marker autoantibody for rheumatoid arthritis: colocalisation of the specific for rheumatoid arthritis but commonly occurs during syno-
perinuclear factor and profilaggrin. Ann Rheum Dis 50:611–618, vitides. J Immunol 174:5057–5064, 2005.
1991. 50. Makrygiannakis D, et al: Citrullination is an inflammation-dependent
24. Janssens X, Veys EM, Verbruggen G, et al: The diagnostic signifi- process. Ann Rheum Dis 65:1219–1222, 2006.
cance of the antiperinuclear factor for rheumatoid arthritis. J Rheu- 51. Makrygiannakis D, et al: Smoking increases peptidylarginine deimi-
matol 15:1346–1350, 1988. nase 2 enzyme expression in human lungs and increases citrullination
25. Cassani F, et al: Antiperinuclear factor in an Italian series of patients in BAL cells. Ann Rheum Dis 67:1488–1492, 2008.
with rheumatoid arthritis. Ric Clin Lab 13:347–352, 1983. 52. Dyson HJ, Wright PE: Antigenic peptides. FASEB J 9:37–42, 1995.
26. Berthelot JM, Maugars Y, Audrain M, et al: Specificity of antiperi- 53. Dorow DS, et al: Two large immunogenic and antigenic myoglobin
nuclear factor for rheumatoid arthritis in rheumatoid factor-positive peptides and the effects of cyclisation. Mol Immunol 22:1255–1264,
sera. Br J Rheumatol 34:716–720, 1995. 1985.
845.e2 PART 7    DIAGNOSTIC TESTS AND PROCEDURES IN RHEUMATIC DIEASES

54. Schellekens GA, et al: The diagnostic properties of rheumatoid 78. Sokolove J, et al: Autoantibody epitope spreading in the pre-clinical
arthritis antibodies recognizing a cyclic citrullinated peptide. Arthritis phase predicts progression to rheumatoid arthritis. PLoS One 7:
Rheum 43:155–163, 2000. e35296, 2012.
55. van Venrooij WJ, Zendman AJ: Anti-CCP2 antibodies: an overview 79. Wagner CA, et al: Identification of anticitrullinated protein anti-
and perspective of the diagnostic abilities of this serological marker body reactivities in a subset of anti-CCP-negative rheumatoid arthri-
for early rheumatoid arthritis. Clin Rev Allergy Immunol 34:36–39, tis: association with cigarette smoking and HLA-DRB1 “shared
2008. epitope” alleles. Ann Rheum Dis 74:579–586, 2015.
56. van Gaalen FA, Visser H, Huizinga TW: A comparison of the diag- 80. van Gaalen FA, et al: Autoantibodies to cyclic citrullinated peptides
nostic accuracy and prognostic value of the first and second anti- predict progression to rheumatoid arthritis in patients with undif-
cyclic citrullinated peptides (CCP1 and CCP2) autoantibody tests ferentiated arthritis: a prospective cohort study. Arthritis Rheum 50:
for rheumatoid arthritis. Ann Rheum Dis 64:1510–1512, 2005. 709–715, 2004.
57. Lutteri L, Malaise M, Chapelle JP: Comparison of second- and third- 81. Jansen LM, et al: The predictive value of anti-cyclic citrullinated
generation anti-cyclic citrullinated peptide antibodies assays for peptide antibodies in early arthritis. J Rheumatol 30:1691–1695, 2003.
detecting rheumatoid arthritis. Clin Chim Acta 386:76–81, 2007. 82. Nell VP, et al: Autoantibody profiling as early diagnostic and prog-
58. dos Anjos LM, et al: A comparative study of IgG second- and third- nostic tool for rheumatoid arthritis. Ann Rheum Dis 64:1731–1736,
generation anti-cyclic citrullinated peptide (CCP) ELISAs and their 2005.
combination with IgA third-generation CCP ELISA for the diagnosis 83. Vencovsky J, et al: Autoantibodies can be prognostic markers of an
of rheumatoid arthritis. Clin Rheumatol 28:153–158, 2009. erosive disease in early rheumatoid arthritis. Ann Rheum Dis 62:427–
59. Masson-Bessière C, et al: The major synovial targets of the rheuma- 430, 2003.
toid arthritis-specific antifilaggrin autoantibodies are deiminated 84. Mewar D, et al: Independent associations of anti-cyclic citrullinated
forms of the alpha- and beta-chains of fibrin. J Immunol 166:4177– peptide antibodies and rheumatoid factor with radiographic severity
4184, 2001. of rheumatoid arthritis. Arthritis Res Ther 8:R128, 2006.
60. Kinloch A, et al: Synovial fluid is a site of citrullination of autoan- 85. Syversen SW, et al: High anti-cyclic citrullinated peptide levels and
tigens in inflammatory arthritis. Arthritis Rheum 58:2287–2295, 2008. an algorithm of four variables predict radiographic progression in
61. Vossenaar ER, et al: Expression and activity of citrullinating pepti- patients with rheumatoid arthritis: results from a 10-year longitudinal
dylarginine deiminase enzymes in monocytes and macrophages. Ann study. Ann Rheum Dis 67:212–217, 2008.
Rheum Dis 63:373–381, 2004. 86. De Rycke L, et al: Rheumatoid factor and anticitrullinated protein
62. Vossenaar ER, et al: Rheumatoid arthritis specific anti-Sa antibodies antibodies in rheumatoid arthritis: diagnostic value, associations with
target citrullinated vimentin. Arthritis Res Ther 6:R142–R150, 2004. radiological progression rate, and extra-articular manifestations. Ann
63. van Beers JJ, et al: Anti-citrullinated fibronectin antibodies in rheu- Rheum Dis 63:1587–1593, 2004.
matoid arthritis are associated with human leukocyte antigen-DRB1 87. Turesson C, et al: Rheumatoid factor and antibodies to cyclic citrul-
shared epitope alleles. Arthritis Res Ther 14:R35, 2012. linated peptides are associated with severe extra-articular manifesta-
64. Darrah E, Rosen A, Giles JT, et al: Peptidylarginine deiminase 2, 3 tions in rheumatoid arthritis. Ann Rheum Dis 66:59–64, 2007.
and 4 have distinct specificities against cellular substrates: novel 88. Quinn MA, et al: Anti-CCP antibodies measured at disease onset
insights into autoantigen selection in rheumatoid arthritis. Ann help identify seronegative rheumatoid arthritis and predict radiologi-
Rheum Dis 71:92–98, 2012. cal and functional outcome. Rheumatology (Oxford) 45:478–480,
65. Harlow L, et al: Identification of citrullinated hsp90 isoforms as novel 2006.
autoantigens in rheumatoid arthritis-associated interstitial lung 89. Kelly CA, et al: Rheumatoid arthritis-related interstitial lung disease:
disease. Arthritis Rheum 65:869–879, 2013. associations, prognostic factors and physiological and radiological
66. Dwivedi N, et al: Felty’s syndrome autoantibodies bind to deiminated characteristics—a large multicentre UK study. Rheumatology (Oxford)
histones and neutrophil extracellular chromatin traps. Arthritis 53:1676–1682, 2014.
Rheum 64:982–992, 2012. 90. Giles JT, et al: Association of fine specificity and repertoire expan-
67. Kinloch A, et al: Identification of citrullinated alpha-enolase as a sion of anticitrullinated peptide antibodies with rheumatoid arthritis
candidate autoantigen in rheumatoid arthritis. Arthritis Res Ther associated interstitial lung disease. Ann Rheum Dis 73:1487–1494,
7:R1421–R1429, 2005. 2014.
68. Matsuo K, et al: Identification of novel citrullinated autoantigens of 91. Aubart F, et al: High levels of anti-cyclic citrullinated peptide auto-
synovium in rheumatoid arthritis using a proteomic approach. Arthri- antibodies are associated with co-occurrence of pulmonary diseases
tis Res Ther 8:R175, 2006. with rheumatoid arthritis. J Rheumatol 38:979–982, 2011.
69. Suzuki A, et al: Anti-citrullinated collagen type I antibody is a target 92. Zhu J, Zhou Y, Chen X, et al: A metaanalysis of the increased risk of
of autoimmunity in rheumatoid arthritis. Biochem Biophys Res rheumatoid arthritis-related pulmonary disease as a result of serum
Commun 333:418–426, 2005. anticitrullinated protein antibody positivity. J Rheumatol 41:1282–
70. Burkhardt H, et al: Humoral immune response to citrullinated col- 1289, 2014.
lagen type II determinants in early rheumatoid arthritis. Eur J 93. Giles JT, et al: Left ventricular structure and function in patients with
Immunol 35:1643–1652, 2005. rheumatoid arthritis, as assessed by cardiac magnetic resonance
71. Okazaki Y, et al: Identification of citrullinated eukaryotic translation imaging. Arthritis Rheum 62:940–951, 2010.
initiation factor 4G1 as novel autoantigen in rheumatoid arthritis. 94. Marasovic-Krstulovic D, Martinovic-Kaliterna D, Fabijanic D, et al:
Biochem Biophys Res Commun 341:94–100, 2006. Are the anti-cyclic citrullinated peptide antibodies independent pre-
72. Goeb V, et al: Candidate autoantigens identified by mass spectrom- dictors of myocardial involvement in patients with active rheumatoid
etry in early rheumatoid arthritis are chaperones and citrullinated arthritis? Rheumatology (Oxford) 50:1505–1512, 2011.
glycolytic enzymes. Arthritis Res Ther 11:R38, 2009. 95. Lopez-Longo FJ, et al: Association between anti-cyclic citrullinated
73. van Beers JJ, et al: The rheumatoid arthritis synovial fluid citrulli- peptide antibodies and ischemic heart disease in patients with rheu-
nome reveals novel citrullinated epitopes in apolipoprotein E, matoid arthritis. Arthritis Rheum 61:419–424, 2009.
myeloid nuclear differentiation antigen, and beta-actin. Arthritis 96. Giles JT, et al: Myocardial citrullination in rheumatoid arthritis: a
Rheum 65:69–80, 2013. correlative histopathologic study. Arthritis Res Ther 14:R39, 2012.
74. Saulot V, et al: Presence of autoantibodies to the glycolytic enzyme 97. Bongartz T, et al: Citrullination in extra-articular manifestations of
alpha-enolase in sera from patients with early rheumatoid arthritis. rheumatoid arthritis. Rheumatology (Oxford) 46:70–75, 2007.
Arthritis Rheum 46:1196–1201, 2002. 98. Sokolove J, et al: Brief report: citrullination within the atheroscle-
75. Romero V, et al: Immune-mediated pore-forming pathways induce rotic plaque: a potential target for the anti-citrullinated protein anti-
cellular hypercitrullination and generate citrullinated autoantigens body response in rheumatoid arthritis. Arthritis Rheum 65:1719–1724,
in rheumatoid arthritis. Sci Transl Med 5:209ra150, 2013. 2013.
76. Tutturen AE, Fleckenstein B, de Souza GA: Assessing the citrulli- 99. Gizinski AM, et al: Rheumatoid arthritis (RA)-specific autoantibod-
nome in rheumatoid arthritis synovial fluid with and without enrich- ies in patients with interstitial lung disease and absence of clinically
ment of citrullinated peptides. J Proteome Res 13:2867–2873, 2014. apparent articular RA. Clin Rheumatol 28:611–613, 2009.
77. Bang H, et al: Mutation and citrullination modifies vimentin to a 100. Fischer A, et al: Lung disease with anti-CCP antibodies but not
novel autoantigen for rheumatoid arthritis. Arthritis Rheum 56:2503– rheumatoid arthritis or connective tissue disease. Respir Med 106:
2511, 2007. 1040–1047, 2012.
CHAPTER 56    Autoantibodies in Rheumatoid Arthritis 845.e3

101. Visser K, et al: Pretreatment serum levels of anti-cyclic citrullinated non-coding segments and a new human gene, PADI6. Gene 330:19–
peptide antibodies are associated with the response to methotrexate 27, 2004.
in recent-onset arthritis. Ann Rheum Dis 67:1194–1195, 2008. 125. Nakashima K, et al: Molecular characterization of peptidylarginine
102. Farragher TM, et al: Benefit of early treatment in inflammatory poly- deiminase in HL-60 cells induced by retinoic acid and 1alpha,25-
arthritis patients with anti-cyclic citrullinated peptide antibodies dihydroxyvitamin D(3). J Biol Chem 274:27786–27792, 1999.
versus those without antibodies. Arthritis Care Res (Hoboken) 62: 126. Nakashima K, Hagiwara T, Yamada M: Nuclear localization of pep-
664–675, 2010. tidylarginine deiminase V and histone deimination in granulocytes.
103. Braun-Moscovici Y, et al: Anti-cyclic citrullinated protein antibodies J Biol Chem 277:49562–49568, 2002.
as a predictor of response to anti-tumor necrosis factor-alpha therapy 127. Cherrington BD, Morency E, Struble AM, et al: Potential role for
in patients with rheumatoid arthritis. J Rheumatol 33:497–500, 2006. peptidylarginine deiminase 2 (PAD2) in citrullination of canine
104. Kurki P, Aho K, Palosuo T, et al: Immunopathology of rheumatoid mammary epithelial cell histones. PLoS One 5:e11768, 2010.
arthritis. Antikeratin antibodies precede the clinical disease. Arthritis 128. Arita K, et al: Structural basis for Ca(2+)-induced activation of
Rheum 35:914–917, 1992. human PAD4. Nat Struct Mol Biol 11:777–783, 2004.
105. Arbuckle MR, et al: Development of anti-dsDNA autoantibodies 129. Asaga H, Nakashima K, Senshu T, et al: Immunocytochemical local-
prior to clinical diagnosis of systemic lupus erythematosus. Scand J ization of peptidylarginine deiminase in human eosinophils and neu-
Immunol 54:211–219, 2001. trophils. J Leukoc Biol 70:46–51, 2001.
106. Arbuckle MR, et al: Development of autoantibodies before the clini- 130. Nachat R, et al: Peptidylarginine deiminase isoforms 1-3 are expressed
cal onset of systemic lupus erythematosus. N Engl J Med 349:1526– in the epidermis and involved in the deimination of K1 and filaggrin.
1533, 2003. J Invest Dermatol 124:384–393, 2005.
107. Kolfenbach JR, et al: Autoimmunity to peptidyl arginine deiminase 131. Esposito G, et al: Peptidylarginine deiminase (PAD) 6 is essential for
type 4 precedes clinical onset of rheumatoid arthritis. Arthritis Rheum oocyte cytoskeletal sheet formation and female fertility. Mol Cell
62:2633–2639, 2010. Endocrinol 273:25–31, 2007.
108. Eriksson C, et al: Autoantibodies predate the onset of systemic lupus 132. Foulquier C, et al: Peptidyl arginine deiminase type 2 (PAD-2) and
erythematosus in northern Sweden. Arthritis Res Ther 13:R30, 2011. PAD-4 but not PAD-1, PAD-3, and PAD-6 are expressed in rheuma-
109. Kokkonen H, et al: Antibodies of IgG, IgA and IgM isotypes against toid arthritis synovium in close association with tissue inflammation.
cyclic citrullinated peptide precede the development of rheumatoid Arthritis Rheum 56:3541–3553, 2007.
arthritis. Arthritis Res Ther 13:R13, 2011. 133. Chang X, et al: Localization of peptidylarginine deiminase 4 (PADI4)
110. Rantapaa-Dahlqvist S, et al: Antibodies against cyclic citrullinated and citrullinated protein in synovial tissue of rheumatoid arthritis.
peptide and IgA rheumatoid factor predict the development of rheu- Rheumatology (Oxford) 44:40–50, 2005.
matoid arthritis. Arthritis Rheum 48:2741–2749, 2003. 134. Arita K, et al: Structural basis for histone N-terminal recognition by
111. Kolfenbach JR, et al: A prospective approach to investigating the human peptidylarginine deiminase 4. Proc Natl Acad Sci U S A 103:
natural history of preclinical rheumatoid arthritis (RA) using first- 5291–5296, 2006.
degree relatives of probands with RA. Arthritis Rheum 61:1735–1742, 135. Slade DJ, et al: Protein arginine deiminase 2 binds calcium in an
2009. ordered fashion: implications for inhibitor design. ACS Chem Biol
112. Berglin E, et al: Radiological outcome in rheumatoid arthritis is pre- 10:1043–1053, 2015.
dicted by presence of antibodies against cyclic citrullinated peptide 136. Darrah E, et al: erosive rheumatoid arthritis is associated with anti-
before and at disease onset, and by IgA-RF at disease onset. Ann bodies that activate PAD4 by increasing calcium sensitivity. Sci Transl
Rheum Dis 65:453–458, 2006. Med 5:186ra65, 2013.
113. Majka DS, et al: Duration of preclinical rheumatoid arthritis-related 137. Hill HR, Augustine NH, Jaffe HS: Human recombinant interferon
autoantibody positivity increases in subjects with older age at time gamma enhances neonatal polymorphonuclear leukocyte activation
of disease diagnosis. Ann Rheum Dis 67:801–807, 2008. and movement, and increases free intracellular calcium. J Exp Med
114. Gregersen PK, Silver J, Winchester RJ: The shared epitope hypoth- 173:767–770, 1991.
esis. An approach to understanding the molecular genetics of suscep- 138. Pozzan T, Lew DP, Wollheim CB, et al: Is cytosolic ionized cal-
tibility to rheumatoid arthritis. Arthritis Rheum 30:1205–1213, 1987. cium regulating neutrophil activation? Science 221:1413–1415,
115. van der Helm-van Mil AH, Huizinga TW: Advances in the genetics 1983.
of rheumatoid arthritis point to subclassification into distinct disease 139. Brandolini L, et al: IL-1 beta primes IL-8-activated human neutro-
subsets. Arthritis Res Ther 10:205, 2008. phils for elastase release, phospholipase D activity, and calcium flux.
116. Huizinga TW, et al: Refining the complex rheumatoid arthritis phe- J Leukoc Biol 59:427–434, 1996.
notype based on specificity of the HLA-DRB1 shared epitope for 140. Wong K, Kwan-Yeung L: Sphingosine mobilizes intracellular calcium
antibodies to citrullinated proteins. Arthritis Rheum 52:3433–3438, in human neutrophils. Cell Calcium 14:493–505, 1993.
2005. 141. Andrade F, et al: Autocitrullination of human peptidyl arginine
117. Irigoyen P, et al: Regulation of anti-cyclic citrullinated peptide anti- deiminase type 4 regulates protein citrullination during cell activa-
bodies in rheumatoid arthritis: contrasting effects of HLA-DR3 and tion. Arthritis Rheum 62:1630–1640, 2010.
the shared epitope alleles. Arthritis Rheum 52:3813–3818, 2005. 142. Mechin MC, et al: Deimination is regulated at multiple levels includ-
118. Snir O, et al: Multiple antibody reactivities to citrullinated antigens ing auto-deimination of peptidylarginine deiminases. Cell Mol Life Sci
in sera from patients with rheumatoid arthritis: association with 67:1491–1503, 2010.
HLA-DRB1 alleles. Ann Rheum Dis 68:736–743, 2009. 143. Borders CL Jr, et al: A structural role for arginine in proteins: mul-
119. van der Helm-van Mil AH, et al: The HLA-DRB1 shared epitope tiple hydrogen bonds to backbone carbonyl oxygens. Protein Sci 3:
alleles differ in the interaction with smoking and predisposition to 541–548, 1994.
antibodies to cyclic citrullinated peptide. Arthritis Rheum 56:425– 144. Tarcsa E, et al: Protein unfolding by peptidylarginine deiminase. Sub-
432, 2007. strate specificity and structural relationships of the natural substrates
120. Johansson M, Arlestig L, Hallmans G, et al: PTPN22 polymorphism trichohyalin and filaggrin. J Biol Chem 271:30709–30716, 1996.
and anti-cyclic citrullinated peptide antibodies in combination 145. Mechin MC, et al: The peptidylarginine deiminases expressed in
strongly predicts future onset of rheumatoid arthritis and has a speci- human epidermis differ in their substrate specificities and subcellular
ficity of 100% for the disease. Arthritis Res Ther 8:R19, 2006. locations. Cell Mol Life Sci 62:1984–1995, 2005.
121. Stanford SM, Mustelin TM, Bottini N: Lymphoid tyrosine phospha- 146. Raijmakers R, et al: Experimental autoimmune encephalomyelitis
tase and autoimmunity: human genetics rediscovers tyrosine phos- induction in peptidylarginine deiminase 2 knockout mice. J Comp
phatases. Semin Immunopathol 32:127–136, 2010. Neurol 498:217–226, 2006.
122. Shirai H, Blundell TL, Mizuguchi K: A novel superfamily of enzymes 147. Loos T, et al: Citrullination of CXCL10 and CXCL11 by peptidylar-
that catalyze the modification of guanidino groups. Trends Biochem ginine deiminase: a naturally occurring posttranslational modifica-
Sci 26:465–468, 2001. tion of chemokines and new dimension of immunoregulation. Blood
123. Thompson PR, Fast W: Histone citrullination by protein arginine 112:2648–2656, 2008.
deiminase: is arginine methylation a green light or a roadblock? ACS 148. Mortier A, et al: Posttranslational modification of the NH2-terminal
Chem Biol 1:433–441, 2006. region of CXCL5 by proteases or peptidylarginine deiminases (PAD)
124. Chavanas S, et al: Comparative analysis of the mouse and human differently affects its biological activity. J Biol Chem 285:29750–
peptidylarginine deiminase gene clusters reveals highly conserved 29759, 2010.
845.e4 PART 7    DIAGNOSTIC TESTS AND PROCEDURES IN RHEUMATIC DIEASES

149. Proost P, et al: Citrullination of CXCL8 by peptidylarginine deimi- patients with rheumatoid arthritis using a novel assay for citrullina-
nase alters receptor usage, prevents proteolysis, and dampens tissue tion of fibrinogen. Arthritis Res Ther 16:498, 2014.
inflammation. J Exp Med 205:2085–2097, 2008. 174. Shi J, et al: Autoantibodies recognizing carbamylated proteins are
150. Struyf S, et al: Citrullination of CXCL12 differentially reduces present in sera of patients with rheumatoid arthritis and predict joint
CXCR4 and CXCR7 binding with loss of inflammatory and anti- damage. Proc Natl Acad Sci U S A 108:17372–17377, 2011.
HIV-1 activity via CXCR4. J Immunol 182:666–674, 2009. 175. Wang Z, et al: Protein carbamylation links inflammation, smoking,
151. Hagiwara T, Nakashima K, Hirano H, et al: Deimination of arginine uremia and atherogenesis. Nat Med 13:1176–1184, 2007.
residues in nucleophosmin/B23 and histones in HL-60 granulocytes. 176. Holzer M, et al: Myeloperoxidase-derived chlorinating species induce
Biochem Biophys Res Commun 290:979–983, 2002. protein carbamylation through decomposition of thiocyanate and
152. Cuthbert GL, et al: Histone deimination antagonizes arginine meth- urea: novel pathways generating dysfunctional high-density lipopro-
ylation. Cell 118:545–553, 2004. tein. Antioxid Redox Signal 17:1043–1052, 2012.
153. Hagiwara T, Hidaka Y, Yamada M: Deimination of histone H2A and 177. Shi J, et al: Anti-carbamylated protein (anti-CarP) antibodies pre-
H4 at arginine 3 in HL-60 granulocytes. Biochemistry 44:5827–5834, cede the onset of rheumatoid arthritis. Ann Rheum Dis 73:780–783,
2005. 2014.
154. Wang Y, et al: Human PAD4 regulates histone arginine methylation 178. Shi J, et al: Anti-carbamylated protein antibodies are present in
levels via demethylimination. Science 306:279–283, 2004. arthralgia patients and predict the development of rheumatoid arthri-
155. Li P, et al: PAD4 is essential for antibacterial innate immunity medi- tis. Arthritis Rheum 65:911–915, 2013.
ated by neutrophil extracellular traps. J Exp Med 207:1853–1862, 179. Gan RW, et al: Anti-carbamylated protein antibodies are present
2010. prior to rheumatoid arthritis and are associated with its future diag-
156. Wright PW, et al: ePAD, an oocyte and early embryo-abundant pep- nosis. J Rheumatol 42:572–579, 2015.
tidylarginine deiminase-like protein that localizes to egg cytoplasmic 180. Thiele GM, et al: Malondialdehyde-acetaldehyde adducts and anti-
sheets. Dev Biol 256:73–88, 2003. malondialdehyde-acetaldehyde antibodies in rheumatoid arthritis.
157. Suzuki A, et al: Functional haplotypes of PADI4, encoding citrulli- Arthritis Rheumatol 67:645–655, 2015.
nating enzyme peptidylarginine deiminase 4, are associated with 181. Hill GE, et al: Association of malondialdehyde-acetaldehyde (MAA)
rheumatoid arthritis. Nat Genet 34:395–402, 2003. adducted proteins with atherosclerotic-induced vascular inflamma-
158. Kang CP, et al: A functional haplotype of the PADI4 gene associated tory injury. Atherosclerosis 141:107–116, 1998.
with increased rheumatoid arthritis susceptibility in Koreans. Arthritis 182. Tuma DJ: Role of malondialdehyde-acetaldehyde adducts in liver
Rheum 54:90–96, 2006. injury. Free Radic Biol Med 32:303–308, 2002.
159. Barton A, et al: A functional haplotype of the PADI4 gene associated 183. Niemela O: Aldehyde-protein adducts in the liver as a result
with rheumatoid arthritis in a Japanese population is not associated of ethanol-induced oxidative stress. Front Biosci 4:D506–D513,
in a United Kingdom population. Arthritis Rheum 50:1117–1121, 1999.
2004. 184. Ayala A, Munoz MF, Arguelles S: Lipid peroxidation: production,
160. Burr ML, et al: PADI4 genotype is not associated with rheumatoid metabolism, and signaling mechanisms of malondialdehyde and
arthritis in a large UK Caucasian population. Ann Rheum Dis 69:666– 4-hydroxy-2-nonenal. Oxid Med Cell Longev 2014:360438, 2014.
670, 2010. 185. Tuma DJ, Thiele GM, Xu D, et al: Acetaldehyde and malondialde-
161. Horikoshi N, et al: Structural and biochemical analyses of the human hyde react together to generate distinct protein adducts in the liver
PAD4 variant encoded by a functional haplotype gene. Acta Crystal- during long-term ethanol administration. Hepatology 23:872–880,
logr D Biol Crystallogr 67:112–118, 2011. 1996.
162. Zhao J, Zhao Y, He J, et al: Prevalence and significance of anti- 186. Tuma DJ, Newman MR, Donohue TM Jr, et al: Covalent binding of
peptidylarginine deiminase 4 antibodies in rheumatoid arthritis. acetaldehyde to proteins: participation of lysine residues. Alcohol Clin
J Rheumatol 35:969–974, 2008. Exp Res 11:579–584, 1987.
163. Harris ML, et al: Association of autoimmunity to peptidyl arginine 187. Anderson DR, et al: Unique antibody responses to malondialdehyde-
deiminase type 4 with genotype and disease severity in rheumatoid acetaldehyde (MAA)-protein adducts predict coronary artery disease.
arthritis. Arthritis Rheum 58:1958–1967, 2008. PLoS One 9:e107440, 2014.
164. Bang SY, et al: Peptidyl arginine deiminase type IV (PADI4) haplo- 188. Rolla R, et al: Detection of circulating antibodies against
types interact with shared epitope regardless of anti-cyclic citrulli- malondialdehyde-acetaldehyde adducts in patients with alcohol-
nated peptide antibody or erosive joint status in rheumatoid arthritis: induced liver disease. Hepatology 31:878–884, 2000.
a case control study. Arthritis Res Ther 12:R115, 2010. 189. Vehkala L, et al: Plasma IgA antibody levels to malondialdehyde
165. Suzuki T, et al: PADI4 and HLA-DRB1 are genetic risks for acetaldehyde-adducts are associated with inflammatory mediators,
radiographic progression in RA patients, independent of ACPA obesity and type 2 diabetes. Ann Med 45:501–510, 2013.
status: results from the IORRA cohort study. PLoS One 8:e61045, 190. Darrah E, Andrade F: Editorial: citrullination, and carbamylation,
2013. and malondialdehyde-acetaldehyde! Oh my! Entering the forest of
166. Halvorsen EH, et al: Serum IgG antibodies to peptidylarginine deim- autoantigen modifications in rheumatoid arthritis. Arthritis Rheuma-
inase 4 in rheumatoid arthritis and associations with disease severity. tol. 67:604–608, 2015.
Ann Rheum Dis 67:414–417, 2008. 191. Hassfeld W, et al: Demonstration of a new antinuclear antibody
167. Halvorsen EH, et al: Serum IgG antibodies to peptidylarginine deim- (anti-RA33) that is highly specific for rheumatoid arthritis. Arthritis
inase 4 predict radiographic progression in patients with rheumatoid Rheum 32:1515–1520, 1989.
arthritis treated with tumour necrosis factor-alpha blocking agents. 192. Steiner G, et al: Purification and partial sequencing of the nuclear
Ann Rheum Dis 68:249–252, 2009. autoantigen RA33 shows that it is indistinguishable from the A2
168. Giles JT, et al: Association of cross-reactive antibodies targeting protein of the heterogeneous nuclear ribonucleoprotein complex.
peptidyl-arginine deiminase 3 and 4 with rheumatoid arthritis- J Clin Invest 90:1061–1066, 1992.
associated interstitial lung disease. PLoS One 9:e98794, 2014. 193. Steiner G, Skriner K, Hassfeld W, et al: Clinical and immunological
169. Ferrari-Lacraz S, et al: Contact with stimulated T cells up-regulates aspects of autoantibodies to RA33/hnRNP-A/B proteins—a link
expression of peptidylarginine deiminase 2 and 4 by human mono- between RA, SLE and MCTD. Mol Biol Rep 23:167–171, 1996.
cytes. Eur Cytokine Netw 23:36–44, 2012. 194. Meyer O, et al: Anti-RA 33 antinuclear autoantibody in rheumatoid
170. Khandpur R, et al: NETs are a source of citrullinated autoantigens arthritis and mixed connective tissue disease: comparison with anti-
and stimulate inflammatory responses in rheumatoid arthritis. Sci keratin and antiperinuclear antibodies. Clin Exp Rheumatol 11:473–
Transl Med 5:178ra40, 2013. 478, 1993.
171. Wang Y, et al: Histone hypercitrullination mediates chromatin 195. Skriner K, et al: Anti-A2/RA33 autoantibodies are directed to the
decondensation and neutrophil extracellular trap formation. J Cell RNA binding region of the A2 protein of the heterogeneous nuclear
Biol 184:205–213, 2009. ribonucleoprotein complex—differential epitope recognition in
172. Keefe D, et al: Perforin triggers a plasma membrane-repair response rheumatoid arthritis, systemic lupus erythematosus, and mixed con-
that facilitates CTL induction of apoptosis. Immunity 23:249–262, nective tissue disease. J Clin Invest 100:127–135, 1997.
2005. 196. Nell-Duxneuner V, et al: Autoantibody profiling in patients with
173. Damgaard D, Senolt L, Nielsen MF, et al: Demonstration of extracel- very early rheumatoid arthritis: a follow-up study. Ann Rheum Dis
lular peptidylarginine deiminase (PAD) activity in synovial fluid of 69:169–174, 2010.
CHAPTER 56    Autoantibodies in Rheumatoid Arthritis 845.e5

197. Fritsch R, et al: Characterization of autoreactive T cells to the auto- 217. Rombouts Y, et al: Anti-citrullinated protein antibodies acquire a
antigens heterogeneous nuclear ribonucleoprotein A2 (RA33) and pro-inflammatory Fc glycosylation phenotype prior to the onset of
filaggrin in patients with rheumatoid arthritis. J Immunol 169:1068– rheumatoid arthritis. Ann Rheum Dis 74:234–241, 2015.
1076, 2002. 218. Pratesi F, Tommasi C, Anzilotti C, et al: Deiminated Epstein-Barr
198. Kouskoff V, et al: Organ-specific disease provoked by systemic auto- virus nuclear antigen 1 is a target of anti-citrullinated protein anti-
immunity. Cell 87:811–822, 1996. bodies in rheumatoid arthritis. Arthritis Rheum 54:733–741, 2006.
199. Kouskoff V, et al: A new mouse model of rheumatoid arthritis: organ- 219. Shi J, Sun X, Zhao Y, et al: Prevalence and significance of antibodies
specific disease provoked by systemic autoimmunity. Ryumachi 37: to citrullinated human papilloma virus-47 E2345-362 in rheumatoid
147, 1997. arthritis. J Autoimmun 31:131–135, 2008.
200. Korganow AS, et al: From systemic T cell self-reactivity to organ- 220. Sjostrom L, Laurell L, Hugoson A, et al: Periodontal conditions in
specific autoimmune disease via immunoglobulins. Immunity 10:451– adults with rheumatoid arthritis. Community Dent Oral Epidemiol
461, 1999. 17:234–236, 1989.
201. Maccioni M, et al: Arthritogenic monoclonal antibodies from K/ 221. Kasser UR, et al: Risk for periodontal disease in patients with long-
BxN mice. J Exp Med 195:1071–1077, 2002. standing rheumatoid arthritis. Arthritis Rheum 40:2248–2251, 1997.
202. Schaller M, Burton DR, Ditzel HJ: Autoantibodies to GPI in rheu- 222. Mercado FB, Marshall RI, Klestov AC, et al: Relationship between
matoid arthritis: linkage between an animal model and human rheumatoid arthritis and periodontitis. J Periodontol 72:779–787,
disease. Nat Immunol 2:746–753, 2001. 2001.
203. Kassahn D, Kolb C, Solomon S, et al: Few human autoimmune sera 223. de Pablo P, Chapple IL, Buckley CD, et al: Periodontitis in systemic
detect GPI. Nat Immunol 3:411–412, 2002. rheumatic diseases. Nat Rev Rheumatol 5:218–224, 2009.
204. Schubert D, Schmidt M, Zaiss D, et al: Autoantibodies to GPI and 224. Lundberg K, Wegner N, Yucel-Lindberg T, et al: Periodontitis in
creatine kinase in RA. Nat Immunol 3:411–413, 2002. RA-the citrullinated enolase connection. Nat Rev Rheumatol 6:
205. Matsumoto I, et al: Low prevalence of antibodies to glucose-6- 727–730, 2010.
phosphate isomerase in patients with rheumatoid arthritis and a 225. Rosenstein ED, Greenwald RA, Kushner LJ, et al: Hypothesis: the
spectrum of other chronic autoimmune disorders. Arthritis Rheum humoral immune response to oral bacteria provides a stimulus for the
48:944–954, 2003. development of rheumatoid arthritis. Inflammation 28:311–318,
206. Aho K, et al: Antifilaggrin antibodies within “normal” range predict 2004.
rheumatoid arthritis in a linear fashion. J Rheumatol 27:2743–2746, 226. McGraw WT, Potempa J, Farley D, et al: Purification, characteriza-
2000. tion, and sequence analysis of a potential virulence factor from Por-
207. Zendman AJ, Vossenaar ER, van Venrooij WJ: Autoantibodies to phyromonas gingivalis, peptidylarginine deiminase. Infect Immun 67:
citrullinated (poly)peptides: a key diagnostic and prognostic marker 3248–3256, 1999.
for rheumatoid arthritis. Autoimmunity 37:295–299, 2004. 227. Lundberg K, et al: Antibodies to citrullinated alpha-enolase peptide
208. Kokkonen H, Johansson M, Innala L, et al: The PTPN22 1858C/T 1 are specific for rheumatoid arthritis and cross-react with bacterial
polymorphism is associated with anti-cyclic citrullinated peptide enolase. Arthritis Rheum 58:3009–3019, 2008.
antibody-positive early rheumatoid arthritis in northern Sweden. 228. Wegner N, et al: Peptidylarginine deiminase from Porphyromonas
Arthritis Res Ther 9:R56, 2007. gingivalis citrullinates human fibrinogen and alpha-enolase: implica-
209. van der Helm-van Mil AH, et al: The HLA-DRB1 shared epitope tions for autoimmunity in rheumatoid arthritis. Arthritis Rheum
alleles are primarily a risk factor for anti-cyclic citrullinated peptide 62:2662–2672, 2010.
antibodies and are not an independent risk factor for development 229. Quirke AM, et al: Heightened immune response to autocitrullinated
of rheumatoid arthritis. Arthritis Rheum 54:1117–1121, 2006. Porphyromonas gingivalis peptidylarginine deiminase: a potential
210. van der Helm-van Mil AH, Wesoly JZ, Huizinga TW: Understanding mechanism for breaching immunologic tolerance in rheumatoid
the genetic contribution to rheumatoid arthritis. Curr Opin Rheuma- arthritis. Ann Rheum Dis 73:263–269, 2014.
tol 17:299–304, 2005. 230. Konig MF, Paracha AS, Moni M, et al: Defining the role of Porphy-
211. Kapitany A, et al: Associations between serum anti-CCP antibody, romonas gingivalis peptidylarginine deiminase (PPAD) in rheumatoid
rheumatoid factor levels and HLA-DR4 expression in Hungarian arthritis through the study of PPAD biology. Ann Rheum Dis pii:ann
patients with rheumatoid arthritis. Isr Med Assoc J 10:32–36, 2008. rheumdis-2014-205385, May 26, 2014. [Epub ahead of print].
212. Scally SW, et al: A molecular basis for the association of the HLA- 231. Konig MF, Bingham CO III, Andrade F: PPAD is not targeted as a
DRB1 locus, citrullination, and rheumatoid arthritis. J Exp Med citrullinated protein in rheumatoid arthritis, but remains a candidate
210:2569–2582, 2013. for inducing autoimmunity. Ann Rheum Dis 74:e8, 2015.
213. Zhao X, et al: Circulating immune complexes contain citrulli- 232. Klareskog L, et al: A new model for an etiology of rheumatoid
nated fibrinogen in rheumatoid arthritis. Arthritis Res Ther 10:R94, arthritis: smoking may trigger HLA-DR (shared epitope)-restricted
2008. immune reactions to autoantigens modified by citrullination. Arthritis
214. Clavel C, et al: Induction of macrophage secretion of tumor necrosis Rheum 54:38–46, 2006.
factor alpha through Fcgamma receptor IIa engagement by rheuma- 233. Linn-Rasker SP, et al: Smoking is a risk factor for anti-CCP antibod-
toid arthritis-specific autoantibodies to citrullinated proteins com- ies only in rheumatoid arthritis patients who carry HLA-DRB1
plexed with fibrinogen. Arthritis Rheum 58:678–688, 2008. shared epitope alleles. Ann Rheum Dis 65:366–371, 2006.
215. Sokolove J, Zhao X, Chandra PE, et al: Immune complexes contain- 234. Klareskog L, Malmstrom V, Lundberg K, et al: Smoking, citrullina-
ing citrullinated fibrinogen costimulate macrophages via Toll-like tion and genetic variability in the immunopathogenesis of rheuma-
receptor 4 and Fcgamma receptor. Arthritis Rheum 63:53–62, 2011. toid arthritis. Semin Immunol 23:92–98, 2011.
216. Rombouts Y, et al: Extensive glycosylation of ACPA-IgG variable 235. Svard A, Kastbom A, Reckner-Olsson A, et al: Presence and utility
domains modulates binding to citrullinated antigens in rheumatoid of IgA-class antibodies to cyclic citrullinated peptides in early rheu-
arthritis. Ann Rheum Dis pii:annrheumdis-2014-206598, Jan 13, matoid arthritis: the Swedish TIRA project. Arthritis Res Ther 10:R75,
2015. [Epub ahead of print]. 2008.
CHAPTER 57 

Acute Phase Reactants and the


Concept of Inflammation
César E. Fors Nieves • Bruce N. Cronstein • Amit Saxena

KEY POINTS responses have the potential to cause significant harm to


the host through processes such as autoimmune diseases,
Inflammation comprises a complex and highly variable set of allergic reactions, and septic shock.
processes that represent a response to tissue damage from
The concept of inflammation itself has evolved over
infection or injury.
thousands of years. Celsus first described the cardinal signs
The acute phase response, a major accompaniment of of redness, swelling, heat, and pain in the first century ad.
inflammation, is induced by inflammation-associated The fifth sign, loss of function, is often attributed to Galen’s
cytokines and includes a reorchestration of acute phase work in second century.1 Since that time, countless advances
protein synthesis by the liver.
have been made in the understanding of inflammation, from
The erythrocyte sedimentation rate (ESR), the most histologic findings of inflammatory cells within tissues, to
commonly used clinical measure of inflammation, depends the discovery of hematologic and soluble mediators such as
on numerous physical and chemical characteristics of blood, cytokines and complement. Also, the molecular signaling
many of which are not related to inflammation. pathways that drive both its protective effects and the inap-
The quintessential acute phase protein, C-reactive protein propriate injurious responses have been recently elucidated.
(CRP), not only offers biomarker utility in the clinic, but also With increasing insight into mechanisms of the inflam-
plays a role in host defense by recognition of biologic matory response has come an appreciation of its significant
substrates, activation of the complement pathway, and by complexity. Molecular and microscopic processes are differ-
binding to leukocytes. ent during acute and chronic stages, and diverse responses
Cytokines, chemokines, adhesion molecules, and other are induced by various types of exogenous and endogenous
products of activated inflammatory cells are secreted during stimuli (e.g., bacteria, viruses, parasites, crystals, allergens,
inflammation and play roles in the inflammatory response, and ischemia). More recently, the role of the milieu of the
but several problems limit the clinical usefulness of their inflammatory response, especially at the level of the vascu-
quantitation for routine clinical purposes. lar endothelium, has taken on considerable importance.2
CRP and ESR may reflect disease activity and correlate with Furthermore, inherent redundancies of functions have been
disease prognosis in rheumatoid arthritis but generally are noted, as have interactions between the mediators of inflam-
not helpful for differential diagnosis. mation that allow for a broad, effective response, but these
Although CRP and ESR correlate with clinical activity in many redundancies make it difficult to understand the pathways
inflammatory rheumatic diseases, the absent or modest CRP of inflammation in a linear fashion. These intricacies have
response seen in some patients with active systemic lupus resulted in a vague definition and on continued reliance on
erythematosus remains unexplained. the final downstream macroscopic cardinal signs to tie
CRP or ESR is elevated in more than 80% of patients with together all of the ongoing processes. It is these same intri-
polymyalgia rheumatica and in approximately 95% of cacies that have made evaluation of inflammation through
patients with giant cell arteritis. Both are useful for disease laboratory tests imprecise.
follow-up, but their imperfect correlation with disease Basic hematologic abnormalities may give clues to the
activity indicates that these measures cannot supplant sound presence of inflammation, but different patterns are often
clinical judgment. associated with underlying causes and are not universally
Minor elevations of CRP, within the normal population found. Leukocytosis can be seen in infections, acute crystal
reference range, are associated with increased risk of diseases, and some autoimmune disorders such as adult
myocardial infarction but are nonspecific (especially in Still’s disease. Anemia is often associated with certain dis-
patients with rheumatic disease). Minor CRP elevation is eases that cause chronic inflammation, such as rheumatoid
associated with well-recognized risk factors for cardiovascular arthritis (RA). Reactive thrombocytosis occurs secondary
disease, which may explain its predictive value. to the release of cytokines after an inciting infectious or
inflammatory event, and the role of platelets and platelet-
derived mediators in stimulating inflammation has been
The inflammatory response is the body’s natural defense described at the molecular level.3
against unchecked tissue damage from infection or injury. Laboratory tests most commonly used by physicians to
It occurs during the acute phase of an inciting event and, obtain information about the extent of inflammation are
if the stimulus is not eliminated, in a chronic, putatively those that measure the acute phase reaction. In response
healing stage. When excessive or uncontrolled, these to injury, local inflammatory cells secrete cytokines that

846
CHAPTER 57    Acute Phase Reactants and the Concept of Inflammation 847

influence the liver to increase or decrease production of TABLE 57-1  Human Acute Phase Proteins
various proteins. The erythrocyte sedimentation rate (ESR) Plasma Proteins that Increase in Inflammation
has been the classic marker of inflammation, with serum Complement System
C-reactive protein (CRP) taking on an increasingly promi-   C3
nent role. Other novel inflammatory markers such as pro-   C4
  C9
calcitonin have been recognized, but their clinical utility  Factor B
has yet to be fully determined.   C1 inhibitor
CRP elevations, especially minor ones, have been noted   C4b-binding protein
in numerous conditions that traditionally have not been  Mannose-binding lectin
considered inflammatory, most significantly involving the Coagulation and fibrinolytic system
cardiovascular system. This has shed light on subclinical  Fibrinogen
inflammation as a possible factor in the pathogenesis of a   Plasminogen
  Tissue plasminogen activator
great number of diseases.  Urokinase
  Protein S
  Vitronectin
ACUTE PHASE RESPONSE   Plasminogen-activator inhibitor 1
Within minutes of tissue injury, activation of the innate Anti-proteases
immune system induces cytokine production that results in   α-Protease inhibitor
a multisystem acute phase response involving the liver, vas-   α-Anti-chymotrypsin
  Pancreatic secretory trypsin inhibitor
cular system, bone marrow, and central nervous system.4,5   Inter-α-trypsin inhibitors
Many elements of the reaction can be regarded as part of
the innate response and are defensive or adaptive in nature.6 Transport proteins
  Ceruloplasmin
Mouse studies have shown that as much as 7% of the regula-   Haptoglobin
tory gene pool undergoes significant changes in expression   Hemopexin
during inflammation, and that induction of liver acute Participants in inflammatory responses
phase genes is mediated by the transcription factor signal  Secreted phospholipase A2
transducer and activator of transcription 3 (STAT3).7-9  Lipopolysaccharide-binding protein
Although the acute phase response can trigger numerous   Interleukin-1–receptor antagonist
neuroendocrine, hematopoietic, and metabolic effects, the   Granulocyte colony-stimulating factor
changes in plasma proteins synthesized by hepatocytes are Others
monitored as signs of underlying inflammation (Tables 57-1   C-reactive protein
 Serum amyloid A
and 57-2). An acute phase protein is one in which the   α-Acid glycoprotein
plasma concentration changes from baseline by at least 25%  Fibronectin
during inflammation; responses vary in terms of concentra-  Ferritin
tion and kinetics (Figure 57-1).10 CRP and serum amyloid   Angiotensinogen
A (SAA) levels increase more than 1000-fold during acute Plasma Proteins that Decrease in Inflammation
infection and peak at 2 to 3 days. Concentrations of other Albumin
proteins peak at longer periods and can range from a 50% Transferrin
Transthyretin
increase in complement and ceruloplasmin, to a several-fold α-HS glycoprotein
amplification in haptoglobin, fibrinogen, α1-proteinase Alpha-fetoprotein
inhibitor, and α1-acid glycoprotein. Other proteins are Thyroxine-binding globulin
negative acute phase proteins, the concentrations of which Insulin-like growth factor I
Factor XII
fall during the inflammatory response. These include anti-
thrombin III, protein S, prealbumin, albumin, transferrin, HS, Heremans-Schmid.
and apolipoprotein A-I.4,5 From Gabay C, Kushner I: Acute-phase proteins and other systemic
Hepatic stimulation of acute phase proteins is induced responses to inflammation. N Engl J Med 340:448–454, 1999.
by cytokines released by activated monocytes, macrophages,
neutrophils, natural killer (NK) cells, and endothelial cells
acting at the front lines of the inflammatory response. The and interactions of cytokines.5 The roles of the acute phase
main cytokine influencing the liver is IL-6, once called the proteins themselves will be discussed throughout the chapter
“hepatocyte-stimulating factor.” It likely mediates protein but have been found to include direct involvement in
expression via the Janus kinase (JAK) and STAT3 path- host defense by activation of the complement, proteinase
ways, as well as C/EBP family members and Rel proteins inhibition, and antioxidant activity.14 However, some of the
(nuclear factor-κB [NF-κB]).9,11 During initial stages, IL-1 described in vitro effects of proteins may not be relevant
and TNF synergize with IL-6 and trigger further IL-6 pro- in vivo.
duction, but their roles are limited.12 The soluble IL-6
receptor amplifies IL-6 effects both locally and systemically. Erythrocyte Sedimentation Rate
IL-6 also performs a protective role during disease, inducing
the expression of an IL-1 receptor antagonist.13 Although ESR is an indirect screen for elevated concentra-
Acute phase protein levels are not uniform in their tions of acute phase proteins, it has been the most widely
expression; this is likely related to the underlying patho- used marker of inflammation for almost a century. Measure-
physiologic state and is regulated by different combinations ment of ESR is performed when blood is placed in a vertical
848 PART 7    DIAGNOSTIC TESTS AND PROCEDURES IN RHEUMATIC DISEASES

TABLE 57-2 Other Acute Phase Phenomena tube and the rate of fall of erythrocytes is measured. The
Neuroendocrine Changes ancient Greeks recognized increased red blood cell (RBC)
Fever, somnolence, and anorexia sedimentation as a way to detect “bad bodily humors,” but
Increased secretion of corticotropin-releasing hormone, our modern understanding and use of RBC sedimentation
corticotropin, and cortisol as a test date back to the German scholar Fahraeus in
Increased secretion of arginine vasopressin
Decreased production of insulin-like growth factor I 1918.15 He determined that certain plasma proteins, espe-
Increased adrenal secretion of catecholamines cially fibrinogen, are able to lower the electrostatic charge
Hematopoeitic Changes
on RBC surfaces so they can aggregate, form rouleaux, and
Anemia of chronic disease fall faster.
Leukocytosis Several factors are involved in acceleration of ESR.
Thrombocytosis Asymmetric plasma proteins, such as fibrinogen and, to a
Metabolic Changes lesser extent, alpha2, beta, and gamma globulins decrease
Loss of muscle and negative nitrogen balance the negative charge of erythrocytes (zeta potential) that
Decreased gluconeogenesis prevents rouleaux formation. Red cell factors also play a role
Osteoporosis
Increased hepatic lipogenesis
in that changes in plasma ratios in anemic states also favor
Increased lipolysis in adipose tissue rouleaux. However, microcytosis, polycythemia, and abnor-
Decreased lipoprotein lipase activity in muscle and adipose tissue mally shaped RBCs (e.g., sickle cells and spherocytes)
Cachexia hinder aggregation and lower the ESR.16 Conditions that
Hepatic Changes elevate fibrinogen, even if they are not necessarily consid-
Increased metallothionein, inducible nitric oxide synthase, heme ered inflammatory, can raise ESR. These include pregnancy,
oxygenase, manganese superoxide dismutase, and tissue inhibitor diabetes, end-stage renal disease, and heart disease. Major
of metalloproteinase-1
Decreased phosphoenolpyruvate carboxykinase activity
increases in the concentration of a single molecular species,
such as a monoclonal immunoglobulin in multiple myeloma,
Changes in Nonprotein Plasma Constituents
Hypozincemia, hypoferremia, and hypercupremia
also cause increased sedimentation.17 The ESR is elevated
Increased plasma retinol and glutathione concentrations in obesity, as is CRP, presumably as a result of IL-6 secretion
by adipocytes.18 Factors such as glucocorticoids, cryoglobu-
From Gabay C, Kushner I: Acute-phase proteins and other systemic lins, hypofibrinogenemia, and hyperviscosity lower the
responses to inflammation. N Engl J Med 340:448–454, 1999. value.4 The physiochemical dynamics that allow for sedi-
mentation has been a continued source of debate, with
disparate models presented to explain how proteins on cell
surfaces interact to cause RBC aggregation.19
Although novel and rapid tests for ESR have proven
30,100 promising, the International Committee for Standardiza-
tion in Hematology continues to recommend the Wester-
30,000
gren technique of testing anti-coagulated blood.20,21 The
700 usual accepted upper limits of normal are 15 mm/hr for
males and 20 mm/hr for females; however, the ESR increases
600 C-reactive protein with age and varies by race, which calls the reliability of the
Plasma concentration

test into question. A simple formula for calculating the


500 upper limit of normal ESR at any age has been used regu-
(% change)

Serum amyloid A larly. In men, age in years divided by 2; in women, 10 plus


400 age in years divided by 2. Despite the ability to control for
300
age, other limitations of the test have been noted and are
Haptoglobin listed in Table 57-3. The relative virtues of CRP determina-
Fibrinogen tion have diminished some of the importance of ESR, but
200
it remains an easy, inexpensive test with a wealth of back-
100 C3 ground literature. Therefore the sedimentation rate contin-
ues to play a prominent role in clinical practice.
0
Albumin Transferrin
C-Reactive Protein
0 7 14 21
C-reactive protein is an acute phase protein, the serum
Inflammatory Days concentration of which reflects ongoing inflammation
stimulus
better than other tests in most, but not all, diseases.22 CRP
Figure 57-1  Typical plasma acute phase protein changes after a mod- was identified in 1930, when sera obtained from patients
erate inflammatory stimulus. Several patterns of response are seen:
major acute phase protein, increase 100-fold (e.g., C-reactive protein, with Streptococcus pneumonia infection were found to
serum amyloid A); moderate acute phase protein, increase twofold to contain a protein that could bind to the “C” polysaccharide
fourfold (e.g., fibrinogen and haptoglobin); minor acute phase protein, of the bacterial cell wall. This protein circulates as a 115 kDa
increase 50% to 100% (e.g., complement C3); and negative acute phase pentamer of noncovalently linked 23 kDa subunits, which
protein, decrease (e.g., albumin, transferrin). (Modified from Gitlin JD,
Colten HR: Molecular biology of the acute-phase plasma proteins. In Pick E,
has been highly conserved during hundreds of millions of
Landy M, editors: Lymphokines, vol 14, San Diego, 1987, Academic Press, years of evolution. In contrast to immunoglobulins and
pp 123–153.) complement components, CRP deficiency in humans has
CHAPTER 57    Acute Phase Reactants and the Concept of Inflammation 849

TABLE 57-3  Comparison of Erythrocyte Sedimentation Rate and C-Reactive Protein


Erythrocyte Sedimentation Rate C-Reactive Protein
Advantages Much clinical information in the literature Rapid response to inflammatory stimuli
May reflect overall health status Wide range of clinically relevant values are detectable
Unaffected by age and gender
Reflects value of a single acute phase protein
Can be measured on stored sera
Quantitation is precise and reproducible
Disadvantages Affected by red blood cell morphology Not sensitive to changes in SLE disease activity
Affected by anemia and polycythemia
Reflects levels of many plasma proteins, not all of which are
acute phase proteins
Responds slowly to inflammatory stimuli
Requires fresh sample
May be affected by drugs

not been described. Genome-wide associated studies per- TABLE 57-4  Conditions Associated with Elevated C-Reactive
formed recently have shown that at least seven distinct loci Protein Levels
are involved in the basal expression of CRP,23-25 which is Normal or Minor Elevation (<1 mg/dL)
upregulated upon stimulation by the transcription factors C/ Vigorous exercise
EBP and Rel.26 It is present in trace concentrations in the Common cold
plasma of all humans (roughly 1 mg/L, with higher concen- Pregnancy
Gingivitis
trations in women and the elderly). Plasma C-reactive Seizures
protein is synthesized by hepatocytes, although other sites Depression
of local production and possibly minimal secretion have Insulin resistance and diabetes
been suggested. Several genetic polymorphisms
Obesity
The precise function of CRP is unknown and may be
varied, but it exhibits important recognition and activation Moderate Elevation (1 to 10 mg/dL)
Myocardial infarction
capabilities, and it binds to numerous ligands.27 CRP recog- Malignancies
nizes phosphocholine, phospholipids, fibronectin, chroma- Pancreatitis
tin, and histones, all of which are exposed at sites of tissue Mucosal infection (bronchitis, cystitis)
damage and by apoptotic cells; CRP may target them for Most systemic autoimune diseases
clearance.28 C-reactive protein bridges the gap between Rheumatoid arthritis
innate and adaptive immunity by activating the classic Marked Elevation (>10 mg/dL)
complement pathway and interacting with cells of the Acute bacterial infection (80% to 85%)
Major trauma
immune system through binding of Fcγ receptors.29,30 CRP Systemic vasculitis
induces inflammatory cytokines, tissue factors, and shedding
of the IL-6 receptor, all of which result in a complement-
dependent increase in tissue damage.28 Other CRP func-
tions are anti-inflammatory, including promotion of the tions greater than 10 mg/dL show marked increases. Most
non-inflammatory clearance of apoptotic cells and preven- patients with extremely high levels (e.g., >15 mg/dL) have
tion of neutrophil adhesion to the endothelium.31,32 Thus bacterial infection. One study found that in patients with
CRP may play many pathophysiologic roles during the CRP concentrations greater than 50 mg/dL, infection was
course of the inflammatory process.14,33 present in 88% of participants.37 Clinical conditions associ-
After an acute inflammatory stimulus, CRP concentra- ated with varying degrees of elevation of CRP are listed in
tion increases rapidly and peaks at 2 to 3 days at levels that Table 57-4, and the range of CRP concentrations in many
reflect the extent of tissue injury. If the stimulus has been rheumatologic diseases is shown in Figure 57-2.
removed, serum CRP levels drop rapidly, with a half-life of Several limitations associated with the use of C-reactive
roughly 19 hours.34 Persistent elevations in CRP are seen in protein measurement must be acknowledged. No unifor-
chronic inflammatory states such as active RA, pulmonary mity in reporting concentrations has been noted between
tuberculosis, or extensive malignant disease. laboratories, and values can be conveyed in mg/L, µg/mL,
Immunoassays and laser nephelometry are used at modest or mg/dL. Similar to ESR, population studies show a skewed,
cost to quantify serum CRP levels. Most healthy adults have rather than gaussian, distribution, which renders parametric
levels less than 0.3 mg/dL. The significance of minor eleva- statistical tests inappropriate for interpretation of CRP
tions in CRP is a subject of debate and will be discussed data. Population differences in CRP levels in the United
subsequently. However, usual methods of CRP determina- States have been reported between sexes and among
tion are less precise at concentrations in the range of 0.3 to racial groups. This is presumably also prevalent as an issue
1 mg/dL, so high-sensitivity (hs) CRP methods are used to in practice in other populations globally. Elevation of
accurately measure these levels. Generally, concentrations C-reactive protein in the elderly may represent age-related
greater than 1 mg/dL reflect clinically significant inflamma- disorders, the pathogenesis of which may involve low-grade
tory disease.35,36 Concentrations of 1 to 10 mg/dL can be inflammation, which complicates the issue of what levels
considered to represent moderate increases, and concentra- are considered normal.38
850 PART 7    DIAGNOSTIC TESTS AND PROCEDURES IN RHEUMATIC DISEASES

Serum CRP Levels (mg/dL)


0.1 0.2 0.5 1.0 2.0 5.0 10 20 50 100

Normal

Osteoarthritis
Rheumatoid
arthritis
Gout
Lupus without
serositis
Lupus with
serositis
Spondylitis
Vasculitis
Adult Still’s
disease

Figure 57-2  Range of C-reactive protein (CRP) levels in rheumatic disease. Authors’ estimates of expected levels of CRP (mg/dL) in certain rheumatic
diseases.

Procalcitonin worth noting that in patients with severe renal dysfunction


these rates may be prolonged, but accumulation does not
Procalcitonin (PCT), a propeptide of calcitonin, has occur.50
recently received increased attention as a useful measure to As a biomarker for infection in autoimmune diseases, the
differentiate between acute bacterial infection and other role of PCT has not yet been fully established. In contrast
inflammatory and febrile syndromes. Studies have shown to CRP, PCT levels are not elevated in in the majority of
that elevated serum levels of PCT have a significantly cases of noninfectious inflammation or nonbacterial infec-
higher specificity for infection than ESR and CRP.39-42 PCT tion. However, exceptions to this rule include some vascu-
has been studied extensively as a clinical tool to facilitate litis syndromes such as Kawasaki’s disease, Goodpasture’s
the decision as to when to start and stop anti-bacterial syndrome, adult-onset Still’s disease51,52 and granulomatosis
agents in patients with pneumonia,43,44 and it has also shown with polyangiitis,53 for which observational studies have
promise in differentiating between infectious and noninfec- demonstrated elevated levels of PCT in patients without
tious causes of fever after orthopedic surgery, in which ESR evidence of bacterial infection. In systemic lupus erythema-
and CRP levels can be difficult to interpret.45,46 For all of tosus, data are mixed, and no definitive statement regarding
these reasons, it remains an intriguing clinical tool for use the usefulness of PCT can be made at this time.54-56
in patients with fevers in the setting of autoimmune disease
who are commonly on immunosuppressive medications and Other Acute Phase Proteins
at higher risk of infection.
Normally produced in the C-cells of the thyroid gland, Measurement of other acute phase proteins has been of
PCT is usually cleaved into calcitonin, katacalcin, and an limited value clinically because their responses to tissue
N-terminal residue. Under normal circumstances, PCT is injury are often slower, and the magnitude of concentration
not released into the bloodstream; however, during severe change is smaller than with CRP. Serum amyloid A (SAA),
infection plasma, levels can increase dramatically.47,48 Inter- a circulating family of proteins produced by hepatocytes,
estingly, this does not lead to an increase in plasma calcito- adipocytes, macrophages, and fibroblast-like synoviocytes
nin levels or activity. At this time, the exact site of PCT has been correlated with disease activity in a number
production, as well as its pathophysiologic role during sepsis, of inflammatory disorders.57 However, reliable testing for
remains uncertain.49 acute phase SAA is not widely available, and data about
In healthy humans, PCT levels are undetectable or very levels expected in disease are limited. Serum ferritin is mod-
low (<0.1 ng/mL). During systemic bacterial infection, erately increased, triggered by cytokines such as IL-1, IL-6,
PCT levels can increase to higher than 100 ng/mL. Cur- IL-18, and TNF. Levels are frequently high in adult-onset
rently available diagnostic tests measure the calcitonin/N- Still’s disease and systemic lupus erythematosus (SLE), and
ProCT part of the protein and therefore only a fragment of they correlate with disease activity.58,59 Hepcidin, a liver-
the 114 to 116 amino-acid chain of the prohormone. A derived anti-microbial peptide and regulator of iron homeo-
useful reference range to exclude sepsis and systemic inflam- stasis, is induced by inflammation, and by IL-6 in particular.60
mation is less than or equal to 0.2 ng/mL. Plasma levels of Its levels rise in parallel with ferritin, and it is important in
greater than or equal to 0.5 ng/mL should be interpreted as the development of anemia of chronic disease, acting as a
abnormal and suggest sepsis. After reaching peak levels, negative regulator of iron absorption and macrophage iron
circulating PCT has a half-life of approximately 22 to 35 release teleologically, to deprive microbes of iron.61 Trans-
hours in serum, and its concentration declines with a 50% ferrin, which binds and transports iron, is a negative acute
plasma-disappearance rate of roughly 1 to 1½ days. It is phase protein.
CHAPTER 57    Acute Phase Reactants and the Concept of Inflammation 851

Apolipoprotein A-I, the principal protein constituent of lives, the presence of blocking factors and natural inhibi-
high-density lipoprotein (HDL), is another negative acute tors, and other technical considerations.71 At present, high
phase protein. In chronic inflammatory diseases such as RA costs, limited availability, and the absence of standardiza-
and SLE, decreased levels may contribute to increased risk tion discourage the measurement of plasma cytokines and
of thrombotic events.62,63 Serum albumin and prealbumin their receptors in clinical practice.
(transthyretin) are also negative acute phase proteins,
although their measurement is not more helpful in diagnosis ACUTE PHASE REACTANTS IN THE
or prognosis than standard tests.64 Serum complement frac- MANAGEMENT OF RHEUMATIC DISEASES
tions become depressed when the system is activated in
certain autoimmune disorders but otherwise rise during the Measurement of ESR and CRP has no role in the diagnosis
acute phase response. of any particular disease, including RA, osteoarthritis, SLE,
PMR, GCA, or other inflammatory arthropathies. However,
these measurements can be clinically helpful in three ways:
Cytokines
(1) in evaluating the extent or severity of inflammation, (2)
Although not acute phase proteins in the classic sense, in monitoring changes in disease activity over time, and (3)
cytokines display the most striking acute phase behavior of in assessing prognosis.
any circulating proteins. IL-6 responds dramatically to tissue
injury, with concentration changes that are faster and Rheumatoid Arthritis
greater than those of CRP or SAA. Acute inflammation and
chronic inflammation have been associated with increases ESR and CRP cannot be used in the diagnosis of RA,
in IL-6, and serum levels of this cytokine have been cor- because 45% of patients may have normal serum levels at
related with the severity and course of disease in RA, presentation,72 although these values represent part of the
juvenile arthritis, ankylosing spondylitis, and polymyalgia diagnostic syndrome or classification criteria sets. These
rheumatica (PMR).65,66 It is also more sensitive than ESR tests are more appropriately applied in RA for monitoring
for detecting disease activity in giant cell arteritis (GCA).67 disease activity and response to therapy. Although ESR
IL-6 levels may be useful in monitoring inflammation if traditionally has been more widely used for these purposes,
hepatocytes are damaged to the point that they are not able many studies have suggested that CRP levels correlate
to synthesize acute phase proteins.4 Knowledge of soluble better with disease activity.73 Some recent reports state that
IL-6 receptor levels may also be helpful in this regard. The CRP levels may overestimate disease response compared
importance of cytokines such as TNF and IL-1 has been with ESR; others claim that differences between the two are
inferred by the successful reduction of inflammation by their minimal.73-75 The existence of patients with depressed CRP
therapeutic inhibitors. Certain diseases, such as the TNF concentrations caused by carrying low-CRP–associated
receptor–associated periodic syndrome (TRAPS) and the genetic variants must be taken into account when this test
autoinflammatory syndromes that involve mutations of the is used universally.76 Matrix metalloproteinase (MMP)-3,
inflammasome controlling IL-1, point to the importance of pro-MMP-3, and soluble E-selectin have also been proposed
these cytokines. as markers for RA disease activity. Their measurements cor-
Increased levels of several other cytokines and circulat- relate with CRP levels but do not provide more information
ing cytokine receptors have been associated with inflamma- than is provided by standard tests.77-79
tion or disease activity as well (Table 57-5).67,68 Different CRP levels average 2 to 3 mg/dL in adult patients with
patterns of cytokine responses have been reported in differ- RA who have moderate disease activity.80 However, varia-
ent diseases, suggesting that cytokine determinations are tion is considerable. At least 5% to 10% of patients have
potentially useful clinically.69,70 However, their quantitation values in the normal range, whereas a few patients with
presents several problems related to their short plasma half- severe disease activity have levels greater than 10 mg/dL.
ESR values have been found to remain stable over the
TABLE 57-5  Products of Inflammatory, Endothelial, and years.81 ESR and CRP have long been used to follow the
Resident Target Tissue Cells/Matrix response to therapy; in general, effective disease-modifying
anti-rheumatic drug therapy decreases CRP levels by
Cytokines and Related Molecules approximately 40%. Inhibition of joint damage by these
Cytokines:
  IL-1
agents usually is accompanied by marked improvement in
  IL-6 acute phase reactants. Progression of joint damage can
  IL-12 occur while the patient is on therapy, however, despite
  IFN-α decreases in ESR and CRP.82 Even more striking improve-
  TNF ment has been seen with biologic agents introduced since
Granulocyte-macrophage colony-stimulating factor
IL-1 receptor antagonist the 1990s, providing objective laboratory support for the
encouraging clinical responses observed. In early reports of
Products of Inflammatory and Endothelial Cells
Calprotectin anti-TNF therapy, CRP and SAA levels declined by 75%
von Willebrand factor and 85%, respectively, in approximately 1 week.83 Treat-
Soluble adhesion molecules (e.g., sVCAM and sE-selectin) ment with abatacept, the T cell CD80/CD86:CD28
Hyaluronic acid co-stimulation modulator, resulted in significant decreases
Collagen and aggrecan degradation products
Osteocalcin
in CRP at both 90 and 360 days of therapy.84 Tofacitinib, a
JAK inhibitor, also appeared to effectively decrease DAS28-
sVCAM, Soluble vascular cell adhesion molecule. ESR scores in RA patients in clinical trials; therefore it may
852 PART 7    DIAGNOSTIC TESTS AND PROCEDURES IN RHEUMATIC DISEASES

be a useful tool to monitor disease activity.85 In one study, Although the concomitant decrease in SAA may point
failure to suppress CRP levels 2 weeks after initiation of against it, several investigations have raised the possibility
infliximab therapy identified most patients who would prove that low CRP levels may be related to the pathogenesis of
to be clinical nonresponders after 12 weeks.86 In contrast to SLE: (1) An association has been noted between SLE and
traditional disease-modifying anti-rheumatic drugs, TNF a genetic polymorphism associated with low CRP levels;
inhibitors have been found to inhibit joint damage even (2) it has been observed that low CRP levels may contrib-
while clinical activity, reflected by CRP levels, remains ute to defective clearance of autoantigens during apopto-
high.87 Tocilizumab, a human IL-6 receptor antibody, sis; and (3) the therapeutic efficacy of CRP has been
improves RA by inhibiting effects of the cytokine. However, reported in mouse models of SLE.101,108-111 Recent studies
because of its mechanism of action, inflammatory markers have also raised the possibility that type I interferon (IFN),
such as ESR and CRP drop to negative values, so that track- which is expressed significantly in SLE, may inhibit CRP
ing them may not reflect the actual effect of the drug; care expression.112,113
must be taken when monitoring disease activity during Substantial CRP elevation in SLE patients is more likely
tocilizumab therapy.88,89 to result from superimposed infection than from activation
ESR and CRP also have value as prognostic indicators in of lupus. CRP levels greater than 6 mg/dL in these patients
RA. Elevated acute phase reactant levels are associated with should serve as an impetus to exclude the possibility of
early synovitis and erosions as detected by magnetic reso- infection, just as they should in other diseases.10 Such levels
nance imaging, with inflammatory cellular infiltrates in should not be regarded as proof of infection, however; as
synovium, and with osteoclastic activation and reduced indicated earlier, marked CRP elevation related to active
bone mineral density.90-92 CRP predicts radiographic pro- SLE can be seen in the absence of infection.
gression, as do ESR and the matrix metalloproteinases Carotid plaque and intima-media wall thickness, corre-
MMP-3 and MMP-1.77,78,93-95 Finally, and perhaps most im- lates of atherosclerotic vascular disease, have been found in
portant, acute phase reactants correlate with work disability association with minor CRP elevation in women with SLE,
on long-term follow-up and predict progression to major as they have in patients with RA.114,115
joint replacement.96,97 As in the normal population, CRP
levels are associated with death from cardiovascular disease.98 Polymyalgia Rheumatica and Giant
In patients with RA in whom heart failure developed, ESR Cell Arteritis
was higher during the 6-month period immediately preced-
ing the onset of heart failure than earlier in tits course.99 The diagnosis of PMR or GCA is supported by an elevated
Serum or synovial fluid levels of many other tissue prod- ESR, often greater than 100 mm/hr. However, such eleva-
ucts (see Table 57-5) have been correlated with clinical tion is no longer regarded as a sine qua non of these disor-
measures of disease activity, severity, and radiographic ders; continuing reports suggest that 10% to 20% of patients
damage. with PMR can have “normal” ESRs, depending on which
Newer quantitative and objective assays that incorporate value is taken as the limit of normal. Such patients tend to
some of the measures previously mentioned as well as other have fewer systemic symptoms and less severe, less frequent
novel ones, have recently come into the market to aid in anemia.116 They have the same frequency of positive tem-
the management and earlier diagnosis of patients with RA. poral artery biopsy results, however, as patients with ele-
Multibiomarker disease activity (MBDA) tests are increas- vated ESR.117,118
ingly popular and could play a significant role the future Only approximately 5% of patients with GCA had ESR
management and study of RA; however, more studies are values less than 40 mm/hr; these patients had fewer visual
required to determine their clinical usefulness across differ- and systemic symptoms than patients with high ESR
ent mechanistic interventions. values.119 In contrast to these findings, ESR and CRP were
found to be significantly lower in patients with ocular
involvement, most commonly in the range of 70 mm/hr to
Systemic Lupus Erythematosus
100 mm/hr. Patients with ESR greater than 100 mm/hr had
Although serum levels of CRP often parallel disease activity decreased incidence of visual ischemic events.120-122
in autoimmune disorders, it has been recognized that SLE In PMR and GCA, CRP and ESR have been regarded
is an exception.100 Although marked CRP responses are in the past as equally valuable in assessing disease activity.
seen in subsets of patients, such as those with serositis or However, recent reports suggest that CRP is more sensitive
chronic synovitis, many (e.g., patients with nephritis) show for both conditions and should be included routinely in the
mild or no elevation during periods of activity.101-103 Serum diagnostic workup.118,123,124 The report that IL-6 is more
levels of SAA are also relatively low in comparison with sensitive than ESR for indicating disease activity in GCA
those of patients with RA, which may explain why rates of is of particular interest.125 Subsets of patients with PMR
secondary amyloidosis are decreased in these patients104 In who have persistently elevated levels of CRP and IL-6
contrast, ESR correlates with disease activity and accrued despite corticosteroid treatment have a higher risk of
tissue damage in SLE.105 Fibrinogen levels increased over relapse.126 A polymorphism at the IL-6 gene promoter has
time in patients, regardless of disease activity.106 Data are characterized these PMR patients with persistently elevated
insufficient to evaluate the potential use of some of the levels of the cytokine.127 Clinical manifestations of disease,
other newer markers described previously, including PCT, even in the presence of a normal ESR or CRP level, should
but many SLE patients with normal CRP levels show ele- not be ignored. Extreme elevation of the ESR in the absence
vated IL-6 concentrations.107 Therefore a deficiency in IL-6 of symptoms of PMR or GCA should raise suspicion of
does not explain the muted CRP response in SLE. other disorders, such as infection, malignancy, or renal
CHAPTER 57    Acute Phase Reactants and the Concept of Inflammation 853

disease. PCT levels in patients with PMR or GCA are Osteoarthritis


usually normal according to a French prospective study.
Elevated PCT levels in these patients with a normal tem- Minor CRP elevations of 0.3 to 1.0 mg/dL have been
poral artery biopsy would be suggestive of an alternate reported in patients with osteoarthritis, particularly those
diagnosis.128 with progressive joint damage.148 However, no evidence
Numerous markers of endothelial perturbation, although supports this association independent of body mass index
not acute phase reactants in the strict sense, are elevated in (BMI), because obesity is a common accompaniment of
plasma in various inflammatory disorders of vessels, particu- osteoarthritis.149 Although local inflammation likely plays a
larly PMR, GCA, and other vasculitides.129 These molecules role in the pathogenesis of osteoarthritis, systemic inflam-
include von Willebrand factor, thrombomodulin, some mation likely does not. However, CRP levels are higher in
vasoactive prostanoids, and a variety of adhesion molecules, patients with erosive osteoarthritis of the hand than in
such as vascular cell adhesion molecule-1. those with nonerosive osteoarthritis.150

Adult-Onset Still’s Disease Other Rheumatic Diseases


Markedly elevated concentrations of ferritin, dispropor- Acute phase markers are elevated in numerous rheumatic
tionately high compared with those of other acute phase diseases while the inflammatory cascade commences. Ele-
reactants, have long been noted in adult-onset Still’s disease vated ESR and CRP can be found in systemic vasculitides,
but are not specific.130 Only a small percentage, commonly crystal arthropathies, psoriatic and reactive arthritides, and
less than 20%, of ferritin is glycosylated in adult-onset infectious joint diseases.151-154 Monitoring for elevated SAA
Still’s disease, a criterion included in recently proposed levels has been proposed as a tool for diagnosis and medica-
classification criteria for this condition.131,132 Extremely tion adjustment in familial Mediterranean fever,155although
elevated ferritin levels have been found in macrophage this test is not widely available, limiting its utility. CRP is
activation syndrome, and 40% of individuals with this con- often normal in patients with primary Sjögren’s syndrome,
dition meet the criteria for adult-onset Still’s disease, sug- and those with elevated responses do not differ clinically
gesting to some that the two disorders are not distinct from patients with normal levels.156 Oligoarticular-onset
entities.133-135 Concentrations of serum IL-18 were extremely juvenile idiopathic arthritis is classically considered as not
elevated in patients with active adult-onset Still’s disease associated with elevated inflammatory markers, although
compared with those of patients with other connective they are present in patients most at risk for systemic
tissue diseases or of healthy individuals and were correlated disease.157
with serum ferritin values and disease severity.136 The cyto-
kine profile in the sera of patients suggests a type 1 T helper PRACTICAL USE OF ACUTE PHASE
(Th) cell response, with significantly higher levels of TNF, REACTANTS
IL-6, and IL-8, in addition to IL-18.137 The role of IL-1
has been inferred from significant improvement in disease In the past, rheumatologists were found to use the ESR more
activity by the IL-1 receptor antagonist, anakinra.138,139 It than twice as frequently as they did CRP levels,158 although
has been suggested that IFN-α may be responsible for the ESR reflects many complex, poorly understood changes in
hyperferritinemia of adult-onset Still’s disease.130 CRP the physical and chemical characteristics of blood not asso-
levels are usually markedly elevated in this disease, and ciated with inflammation. As indicated earlier, the refer-
PCT levels have also been found to be disproportionately ence normal values for ESR are unclear. It is well established
elevated without evidence of acute bacterial infection in that mean ESR values increase substantially with age and
these patients.51,52 differ between men and women. Difficulties associated with
interpretation of the ESR and an increasingly positive clini-
cal experience with CRP suggest that rheumatologists may
Ankylosing Spondylitis
benefit from relying more on CRP than ESR testing.159 No
Ankylosing spondylitis ordinarily does not lead to a substan- single ideal test can be used to evaluate the acute phase
tial increase in ESR or CRP. Median ESR and CRP levels response, however. The relative virtues of these tests in fol-
are 13 mm/hr and 1.6 mg/dL, respectively, in patients with lowing patients with RA have been discussed.74,160
only spinal involvement, and 21 mm/hr and 2.5 mg/dL in Discrepancies between ESR and CRP may result from
patients with peripheral involvement or associated inflam- effects of blood constituents that are not related to inflam-
matory bowel disease.140 Treatment with infliximab led to mation, but that can influence the ESR. In addition,
an average decrease of 75% in CRP concentration after patterns of acute phase protein changes differ in different
12 weeks. Patients with lower CRP levels showed little conditions.5 ESR may be markedly elevated in many patients
improvement, however, raising the possibility that patients with active SLE, whereas the CRP is normal. Undoubtedly,
with higher CRP values show better responses to anti-TNF numerous other clinical situations exist in which similar
treatment than patients with lower CRP levels.141-143 High- discrepancies occur. Although a falsely high ESR has many
sensitivity CRP may better correlate with clinical disease non-inflammatory physicochemical causes (some known,
activity compared with standard CRP testing.144 It has been and many unknown), CRP values greater than 1 mg/dL
reported that levels of IL-8, IL-17, and IL-23 are elevated almost invariably reflect a clinically significant inflamma-
in the serum of patients with active ankylosing spondylitis, tory process. In light of these considerations, many authors
and polymorphisms in the IL-23 receptor gene are associ- believe that several tests, rather than a single test, should
ated with the disease.145-147 be performed and interpreted in their clinical context. It
854 PART 7    DIAGNOSTIC TESTS AND PROCEDURES IN RHEUMATIC DISEASES

has been suggested that ESR, which is associated with explanations have been suggested, including the question
anemia and immunoglobulin levels, may reflect general of the importance of lowering LDL even among patients
severity in RA, whereas CRP is a better test of active inflam- with healthy levels.177 Whether to target CRP levels in
mation per se.161 cardiovascular disease is an ongoing topic of intense debate.
Because no CRP inhibitor drugs are available to directly
C-REACTIVE PROTEIN AND measure the effect of lowering the protein, recent studies
HEALTH: ASSOCIATIONS WITH have centered on observing patients with genetic variations
NONRHEUMATOLOGIC CONDITIONS that result in different baseline levels of CRP. To date,
results have been conflicting with regard to the role
Although most ostensibly healthy individuals have CRP of genetically determined CRP levels in coronary heart
concentrations of 0.3 mg/dL or less, some have concentra- disease.178-182
tions greater than 1 mg/dL. Such minor CRP elevation has Epidemiologic studies that describe an association of
long been attributed to trivial tissue injury or to minimal CRP levels with morbidity and mortality in many chronic
inflammatory processes, such as gingivitis. However, recent diseases, and even in normal aging, have become a cottage
data indicate that CRP concentrations between 0.3 and industry. It is important to remember that these are obser-
1 mg/dL have clinical relevance. This finding has led to an vational population studies. Although such associations
explosion of published literature measuring CRP levels in may have broad and intriguing implications, particularly at
cardiac, neurologic, neoplastic, pulmonary, and even psy- a societal level, they reflect probabilities. This limits their
chiatric disease.162-166 The foundation of these investigations clinical value when they are applied to individual patients.
is the well-established notion that when a high-sensitivity
CRP assay is used, serum CRP levels greater than 0.3 mg/
dL indicate increased the relative risk of atherogenesis and Full references for this chapter can be found on
ExpertConsult.com.
future myocardial infarction.167 The statistical strength of
this is as robust as, but not more robust than, established
risk factors such as hypertension, diabetes, and hyper­
cholesterolemia.168-170 The primary unanswered questions SELECTED REFERENCES
remain: Why does CRP predict myocardial infarction? Is it 2. Biedermann B: Vascular endothelium: checkpoint for inflammation
and immunity. News Physiol Sci 16:84–88, 2001.
a pathogenic mediator itself? 3. Gawaz M, Langer H, May A: Platelets in inflammation and athero-
The observation that CRP is associated with many non- genesis. J Clin Invest 115:3378–3384, 2005.
inflammatory conditions (e.g., low levels of physical activ- 4. Dayer E, Dayer JM, Roux-Lombard P: Primer: the practical use of
ity, low intake of fruits and vegetables, a variety of other biologic markers of rheumatic and systemic inflammatory diseases.
“unhealthy” diets, smoking, hypertension, obesity, sleep Nat Clin Pract Rheum 3:512–520, 2007.
5. Gabay C, Kushner I: Acute-phase proteins and other systemic
deprivation, and low alcohol intake) indicates that classic responses to inflammation. N Engl J Med 340:448–454, 1999.
inflammation does not invariably underlie a CRP response.171 6. Yoo JY, Desiderio S: Innate and acquired immunity intersect in a
Many of these CRP-associated conditions are known to be global view of the acute-phase response. Proc Natl Acad Sci U S A
risk factors for cardiovascular disease, confounding the situ- 100:1157–1162, 2003.
8. Alonzi T, Maritano D, Gorgoni B, et al: Essential role of STAT3 in
ation and suggesting that CRP predicts myocardial infarc- the control of the acute-phase response as revealed by inducible gene
tion because of its association with these risk factors. In activation in the liver. Mol Cell Biol 21:1621–1632, 2001.
addition, elevated CRP may not reflect inflammation, but 10. Morley JJ, Kushner I: Serum C-reactive protein levels in disease. Ann
rather a response to the presence of distressed, metabolically N Y Acad Sci 389:406–418, 1982.
disturbed cells.171 This reverse causation offers a potential 12. Xing Z, Gauldie J, Cox G, et al: IL-6 is an anti-inflammatory cytokine
required for controlling local or systemic acute inflammatory
explanation of the laboratory result, because atherosclerosis responses. J Clin Invest 101:311–320, 1998.
might trigger an elevation in CRP. 13. Jones SA, Horiuchi S, Topley N, et al: The soluble interleukin 6
Nevertheless, CRP has been known for many years to receptor: mechanisms of production and implications in disease.
bind to low-density lipoprotein (LDL) and to activate FASEB J 15:43–58, 2001.
14. Volanakis JE: Human C-reactive protein: expression, structure, and
complement—a potentially pro-inflammatory response; it function. Mol Immunol 38:189–197, 2001.
has also been detected in atherosclerotic plaques.29,172-174 17. Sox HC Jr, Liang MH: The erythrocyte sedimentation rate: guide-
These observations have raised the possibility that CRP lines for rational use. Ann Intern Med 104:515–523, 1986.
may play a direct causal role in coronary heart disease. 18. Bastard JP, Maachi M, Van Nhieu JT, et al: Adipose tissue IL-6
Proatherogenic effects secondary to CRP injections given content correlates with resistance to insulin activation of glucose
uptake both in vivo and in vitro. J Clin Endocrinol Metab 87:2084–
to mice, however, may have been caused by contaminants 2089, 2002.
in commercial CRP preparations, not by CRP itself.175 20. Cha CH, Park CJ, Cha YJ, et al: Erythrocyte sedimentation rate
Because statin drugs lower CRP and LDL, some consider measurements by TEST 1 better reflect inflammation than do those
their effects to provide indirect evidence of a causal role of by the Westergren method in patients with malignancy, autoimmune
disease, or infection. Am J Clin Pathol 131:189–194, 2009.
CRP. In the justification for the use of statins in prevention: 22. Pepys MB, Hirschfield GM: C-reactive protein: a critical update.
an intervention trial evaluating rosuvastatin (JUPITER) J Clin Invest 111:1805–1812, 2003.
trial, patients with normal LDL and elevated CRP were 23. Ridker PM, Pare G, Parker A, et al: Loci related to metabolic-
prescribed rosuvastatin, 20 mg daily, or placebo, and were syndrome pathways including LEPR, HNF1A, IL6R, and GCKR
followed prospectively. Those in the treatment arm had associate with plasma C-reactive protein: the Women’s Genome
Health Study. Am J Hum Genet 82:1185–1192, 2008.
significant reductions in major cardiovascular events.176 26. Cha-Molstad H, Young DP, Kushner I, et al: The interaction of C-rel
However, whether the effects of the study can be solely with C/EBPbeta enhances C/EBPbeta binding to the C-reactive
dependent on CRP reduction remains unclear; alternative protein gene promoter. Mol Immunol 44:2933–2942, 2007.
CHAPTER 57    Acute Phase Reactants and the Concept of Inflammation 855

28. Griselli M, Herbert J, Hutchinson WL, et al: C-reactive protein and 63. Park YB, Lee SK, Lee WK, et al: Lipid profiles in untreated patients
complement are important mediators of tissue damage in acute myo- with rheumatoid arthritis. J Rheumatol 26:1701–1704, 1999.
cardial infarction. J Exp Med 190:1733–1740, 1999. 64. Johnson AM, Merlini G, Sheldon J, et al: Clinical indications for
29. Marnell L, Mold C, Du Clos TW: C-reactive protein: an activator of plasma proteins assays: transthyretin (prealbumin) in inflammation
innate immunity and a modulator of adaptive immunity. Immunol Res and malnutrition—International Federation of Clinical Chemistry
30:261–277, 2004. and Laboratory Medicine (IFCC), IFCC Scientific Division Commit-
31. Gershov D: C-reactive protein binds to apoptotic cells, protects the tee on Plasma Proteins (C-PP). Clin Chem Lab Med 45:419–426,
cells from assembly of the terminal complement components, and 2007.
sustains an anti-inflammatory innate immune response: implications 65. Tutuncu ZN, Bilgie A, Kennedy LG, et al: Interleukin-6, acute phase
for systemic autoimmunity. J Exp Med 192:1353–1364, 2000. reactants and clinical status in anklyosing spondylitis. Ann Rheum
32. Zouki C, Beauchamp M, Baron C, et al: Prevention of in vitro neu- Dis 53:425–426, 1994.
trophil adhesion to endothelial cells through shedding of L-selectin 66. Uddhammar A, Sundqvist KG, Ellis B, et al: Cytokines and adhesion
by C-reactive protein and peptides derived from C-reactive protein. molecules in patients with polymyalgia rheumatica. Br J Rheumatol
J Clin Invest 100:522–529, 1997. 37:766–769, 1998.
34. Vigushin DM, Pepys MB, Hawkins PN: Metabolic and scintigraphic 67. Luqmani R, Sheeran T, Robinson M, et al: Systemic cytokine mea-
studies of radioiodinated human C-reactive protein in health and surements: their role in monitoring the response to therapy in
disease. J Clin Invest 91:1351–1357, 1993. patients with rheumatoid arthritis. Clin Exp Rheumatol 12:503–508,
35. Morley JJ, Kushner I: Serum C-reactive protein levels in disease. Ann 1994.
N Y Acad Sci 389:406–418, 1982. 68. Pountain G, Hazleman B, Cawston TE: Circulating levels of IL-1beta,
36. Macy EM, Hayes TE, Tracy RP: Variability in the measurement of IL-6 and soluble IL-2 receptor in polymyalgia rheumatica and giant
C-reactive protein in healthy subjects: implications for reference cell arteritis and rheumatoid arthritis. Br J Rheumatol 37:797–798,
intervals and epidemiological applications. Clin Chem 43:52–58, 1998.
1997. 69. Gabay C, Cakir N, Moral F, et al: Circulating levels of tumor necrosis
37. Vanderschueren S, Deeren D, Knockaert DC, et al: Extremely ele- factor soluble receptors in systemic lupus erythematosus are signifi-
vated C-reactive protein. Eur J Intern Med 17:430–433, 2006. cantly higher than in other rheumatic diseases and correlate with
38. Woloshin S, Schwartz LM: Distribution of C-reactive protein values disease activity. J Rheumatol 24:303–308, 1997.
in the United States. N Engl J Med 352:1611–1613, 2005. 70. Gabay C, Gay-Croisier F, Roux-Lombard P, et al: Elevated serum
39. Wu JY, Lee SH, Chang SS, et al: Use of serum procalcitonin to levels of interleukin-1 receptor antagonist in polymyositis/
detect bacterial infection in patients with autoimmune diseases: a dermatomyositis: a biologic marker of disease activity with a possible
systemic review and meta-analysis. Arthritis Rheum 64:3034–3042, role in the lack of acute-phase protein response. Arthritis Rheum
2012. 37:1744–1751, 1994.
41. Wacker C, Prkno A, Schlattmann P, et al: Procalcitonin as a diag- 72. Sokka T, Pincus T: Erythrocyte sedimentation rate, C-reactive
nostic marker for sepsis: a systematic review and meta-analysis. Lancet protein, or rheumatoid factor is normal at presentation in 35-45% of
Infect Dis 13:426–435, 2013. patients with rheumatoid arthritis seen between 1980 and 2004:
43. Christ-Crain M, Jaccard-Stolz D, Bingisser R, et al: Effect of analysis from Finland and the United States. J Rheumatol 36:1387–
procalcitonin-guided treatment on antibiotic use and outcome in 1390, 2009.
lower respiratory tract infections: cluster-randomised, single-blinded 73. Matsui T, Kuga Y, Kaneko A, et al: Disease Activity Score 28
intervention trial. Lancet 363:600–607, 2004. (DAS28) using C-reactive protein underestimates disease activity
44. Christ-Crain M, Stolz D, Bissinger R, et al: Procalcitonin guidance and overestimates EULAR response criteria compared with DAS28
of antibiotic therapy in community-acquired pneumonia: a random- using erythrocyte sedimentation rate in a large observational cohort
ized trial. Am J Respir Crit Care Med 174:84–93, 2006. of rheumatoid arthritis patients in Japan. Ann Rheum Dis 66:1221–
47. Snider RH, Nylen ES, Becker KL: Procalcitonin and its component 1226, 2007.
peptides in systemic inflammation: immunochemical characteriza- 76. Rhodes B, Merriman ME, Harrison A, et al: A genetic association
tion. J Investig Med 45:552–560, 1997. study of serum acute-phase C-reactive protein levels in rheumatoid
49. Assicot M, Gendrel D, Carsin H, et al: High serum procalcitonin arthritis: implications for clinical interpretation. PLoS Med 7:
concentrations in patients with sepsis and infection. Lancet 341:515– e1000341, 2010.
518, 1993. 78. Posthumus MD, Limburg PC, Westra J, et al: Serum matrix metal-
51. Chen DY, Chen YM, Lan JL, et al: Diagnostic value of procalcitonin loproteinase 3 levels in comparison to C-reactive protein in periods
for differentiation between bacterial infection and non-infectious with and without progression of radiological damage in patients with
inflammation in febrile patients with active adult-onset Still’s disease. early rheumatoid arthritis. Clin Exp Rheumatol 21:465–472, 2003.
Ann Rheum Dis 68:1074–1075, 2009. 79. Kuuliala A, Eberhardt K, Takala A, et al: Circulating soluble
52. Scire CA, Cavagna L, Montecucco C, et al: Diagnostic value of E-selectin in early rheumatoid arthritis: a prospective five year study.
procalcitonin measurement in febrile patients with systemic autoim- Ann Rheum Dis 61:242–246, 2002.
mune diseases. Clin Exp Rheumatol 24:123–128, 2006. 80. Aletaha D, Smolen JS: The rheumatoid arthritis patient in the clinic:
54. Lanoix JP, Bourgeois AM, Schmidt J, et al: Serum procalcitonin does comparing more than 1300 consecutive DMARD courses. Rheumatol-
not differentiate between infection and disease flare in patients with ogy (Oxford) 41:1367–1374, 2002.
systemic lupus erythematosus. Lupus 20:125–130, 2011. 81. Wolfe F, Pincus T: The level of inflammation in rheumatoid arthritis
55. Kim HA, Jeon JY, An JM, et al: C-reactive protein is a more sensitive is determined early and remains stable over the longterm course of
and specific marker for diagnosing bacterial infections in systemic the illness. J Rheumatol 28:1817–1824, 2001.
lupus erythematosus compared to S100A8/A9 and procalcitonin. 82. Sanmarti R, Gomez A, Ercilla G, et al: Radiological progression in
J Rheumatol 39:728–734, 2012. early rheumatoid arthritis after DMARDS: a one-year follow-up study
56. Bador KM, Intan S, Hussin S, et al: Serum procalcitonin has negative in a clinical setting. Rheumatology (Oxford) 42:1044–1049, 2003.
predictive value for bacterial infection in active systemic lupus ery- 83. Charles P, Elliott MJ, Davis D, et al: Regulation of cytokines, cyto-
thematosus. Lupus 21:1172–1177, 2011. kine inhibitors, and acute-phase proteins following anti-TNF alpha
58. Efthimiou P, Paik PK, Bielory L: Diagnosis and management of adult therapy in rheumatoid arthritis. J Immunol 163:1521–1528, 1999.
onset Still’s disease. Ann Rheum Dis 65:564–572, 2006. 84. Weisman MH, Durez P, Hallegua D, et al: Reduction of inflammatory
59. Nishiya K, Hashimoto K: Elevation of serum ferritin levels as a biomarker response by abatacept in treatment of rheumatoid arthritis.
marker for active systemic lupus erythematosus. Clin Exp Rheumatol J Rheumatol 33:2162–2166, 2006.
15:39–44, 1997. 85. Lee EB, Fleischmann R, Hall S, et al: Tofacitinib versus methotrexate
60. Nemeth E, Valore EV, Territo M, et al: Hepcidin, a putative mediator in rheumatoid arthritis. N Engl J Med 370:2377–2386, 2014.
of anemia of inflammation, is a type II acute-phase protein. Blood 86. Buch MH, Seto Y, Bingham SJ, et al: C-reactive protein as a predic-
101:2461–2463, 2003. tor of infliximab treatment outcome in patients with rheumatoid
62. Lahita RG, Rivkin E, Cavanagh I, et al: Low levels of total choles- arthritis: defining subtypes of nonresponse and subsequent response
terol, high-density lipoprotein, and apolipoprotein A1 in association to etanercept. Arthritis Rheum 52:42–48, 2005.
with anticardiolipin antibodies in patients with systemic lupus ery- 87. Smolen JS, Van Der Heijde DM, St Clair EW, et al: Predictors of
thematosus. Arthritis Rheum 36:1566–1574, 1993. joint damage in patients with early rheumatoid arthritis treated with

You might also like