Seminars in Arthritis and Rheumatism 59 (2023) 152176
Available online 11 February 2023
0049-0172/© 2023 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-
nc-nd/4.0/).
Serum antibodies to periodontal pathogens prior to rheumatoid arthritis
diagnosis: A case-control study
Joyce A. Lee
a
, Ted R. Mikuls
b
,
c
, Kevin D. Deane
d
, Harlan R. Sayles
e
, Geoffrey M. Thiele
b
,
c
, Jess
D. Edison
f
, Brandie D. Wagner
g
, Marie L. Feser
d
, Laura K. Moss
d
, Lindsay B. Kelmenson
d
,
William H. Robinson
h
, Jeffrey B. Payne
a
,
b
,
*
a
Department of Surgical Specialties, Division of Periodontics, College of Dentistry, University of Nebraska Medical Center, Lincoln, NE, USA
b
Department of Internal Medicine, Division of Rheumatology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
c
Medicine, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
d
Department of Internal Medicine, Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
e
Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE USA
f
Department of Internal Medicine, Rheumatology Service, Walter Reed National Military Medical Center, Bethesda, MD, USA
g
Department of Biostatistics and Informatics, School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
h
Geriatrics Research Education and Clinical Center, Veterans Affairs Palo Alto Healthcare System and Division of Immunology/Rheumatology, Stanford University
School of Medicine, Palo Alto, CA, USA
ARTICLE INFO
Keywords:
Rheumatoid arthritis
Periodontitis
ACPA
Rheumatoid factor
Porphyromonas gingivalis
Prevotella intermedia
ABSTRACT
Objectives: 1) To quantify the association between anti-Porphyromonas gingivalis serum antibody concentrations
and the risk of developing rheumatoid arthritis (RA), and 2) to quantify the associations among RA cases between
anti-P. gingivalis serum antibody concentrations and RA-specic autoantibodies. Additional anti-bacterial anti-
bodies evaluated included anti-Fusobacterium nucleatum and anti-Prevotella intermedia.
Methods: Serum samples were acquired pre- and post- RA diagnosis from the U.S. Department of Defense Serum
Repository (n = 214 cases, 210 matched controls). Using separate mixed-models, the timing of elevations of anti-
P. gingivalis, anti-P. intermedia, and anti-F. nucleatum antibody concentrations relative to RA diagnosis were
compared in RA cases versus controls. Associations were determined between serum anti-CCP2, ACPA ne
specicities (vimentin, histone, and alpha-enolase), and IgA, IgG, and IgM RF in pre-RA diagnosis samples and
anti-bacterial antibodies using mixed-effects linear regression models.
Results: No compelling evidence of case-control divergence in serum anti-P. gingivalis, anti-F. nucleatum, and anti-
P. intermedia was observed. Among RA cases, including all pre-diagnosis serum samples, anti-P. intermedia was
signicantly positively associated with anti-CCP2, ACPA ne specicities targeting vimentin, histone, alpha-
enolase, and IgA RF (p<0.001), IgG RF (p = 0.049), and IgM RF (p = 0.004), while anti-P. gingivalis and anti-
F. nucleatum were not.
Conclusions: No longitudinal elevations of anti-bacterial serum antibody concentrations were observed in RA
patients prior to RA diagnosis compared to controls. However, anti-P. intermedia displayed signicant associa-
tions with RA autoantibody concentrations prior to RA diagnosis, suggesting a potential role of this organism in
progression towards clinically-detectable RA.
Introduction
It has been hypothesized that rheumatoid arthritis (RA) may be
initiated in mucosal tissues, including the periodontium [1]. Periodon-
titis (PD) is a biolm-driven inammatory disease of the soft and hard
tissues in the oral cavity resulting from an interaction between the host
immune response and a dysbiotic oral microbiota, ultimately leading to
tooth loss [2]. Over the past few decades, there has been an increased
awareness of the relationship between RA and PD. Both diseases share
similar inammatory pathways and risk factors [3]. Several studies have
* Corresponding author at: Professor and F. Gene and Rosemary Dixon Endowed Chair in Dentistry, Department of Surgical Specialties, College of Dentistry,
Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, 4000 East Campus Loop South, Lincoln, NE, USA.
E-mail address: [email protected] (J.B. Payne).
Contents lists available at ScienceDirect
Seminars in Arthritis and Rheumatism
journal homepage: www.elsevier.com/locate/semarthrit
https://doi.org/10.1016/j.semarthrit.2023.152176
Seminars in Arthritis and Rheumatism 59 (2023) 152176
2
demonstrated PD as a risk factor for RA [46]. It has been speculated
that this relationship may be mediated through the oral periodontal
pathogen, Porphyromonas gingivalis [7].
P. gingivalis is a gram-negative anaerobe recognized as a keystone
pathogen in the pathogenesis of PD [8,9]. Uniquely, it is the only pro-
karyote that can express a functional bacterial peptidyl arginine dei-
minase (PAD) enzyme (often termed PPAD) as a primary virulence
factor, thus serving as a microbe of interest in its role with RA and PD
[10]. The discovery of P. gingivalis PAD led to the hypothesis that
P. gingivalis PAD-mediated protein citrullination at affected periodontal
sites can launch a sequence of events that culminate in the generation of
anti-citrullinated protein antibodies (ACPAs) and, eventually, in the
clinical manifestation of RA [11]. However, while P. gingivalis is a
periodontal pathogen implicated in RA pathogenesis, other bacterial
species involved in PD, such as Prevotella intermedia and Fusobacterium
nucleatum, may also inuence development of RA [12,13].
We hypothesized that circulating concentrations of antibody to
P. gingivalis would be higher in samples from individuals later devel-
oping RA compared to controls. Anti-bacterial serologies may be used as
a surrogate of exposure to periodontal pathogens and we have previ-
ously reported associations between serum antibody to P. gingivalis and
RA-related autoantibody expression among patients without clinically
apparent RA, but with a higher risk of future disease [14]. Moreover, we
postulated that, among those with RA, anti-P. gingivalis antibodies would
be associated with the presence of RA-related autoantibodies prior to
diagnosis. The purpose of this study was to: 1) quantify the association
between anti-P. gingivalis serum antibody concentrations and the risk of
developing RA, and 2) quantify the associations among RA cases be-
tween anti-P. gingivalis serum antibody concentrations and RA-specic
autoantibodies. Additional anti-bacterial antibodies evaluated
included anti-P. intermedia and anti-F. nucleatum to determine whether
associations observed were specic to P. gingivalis or related to a broader
dysbiosis that may be observed in PD.
Methods
Patient population
Study participants consisted of military personnel participating in
the U.S. Department of Defense Serum Repository (DoDSR). Since 1996,
DoDSR has been collecting serum samples to observe health history in
the military population and further understand the risks of deployment
concerning subsequent injuries or chronic illnesses [15].
Active-duty personnel with 2 RA diagnostic codes (1 from a
rheumatologist) were screened from the militarys electronic medical
records [16]. The records were further examined to obtain the date of
diagnosis and fulllment of the 1987 American College of Rheuma-
tology classication criteria [17]. Serum samples were acquired prior to
and after RA diagnosis for up to four samples per case, a minimum of two
samples and up to three samples from pre-diagnosis, collected at
different time points, and one sample from post-diagnosis. This study
utilized 214 RA cases who received a diagnosis of RA between 1995 and
2012. Out of these cases, 212 met the 1987 RA classication criteria and
the other two cases were diagnosed by a board-certied rheumatologist.
These RA cases were chosen because there was a clear date of RA
diagnosis recorded, adequate information to evaluate the clinical course
of their RA after diagnosis, and two or more pre-diagnosis and one
post-diagnosis serum samples with adequate volumes available for
analysis.
Controls were selected and matched to each case based on age (at
time of RA diagnosis for their matched cases), sex, ethnicity, enlistment
region, and duration of sample storage. Exclusions for the controls were
a history of RA or other inammatory arthritis [16].
Four of these controls were subsequently excluded due to insufcient
information available to exclude inammatory arthritis, leaving a total
of 210 controls evaluable for the analysis. These cases and controls were
included in earlier DoDSR studies by our group [16,18].
Clinical data collected included: age at time of diagnosis, sex,
ethnicity, smoking status (those with missing data after chart review
were imputed as never smokers), sample collection timing relative to RA
diagnosis, follow-up time and RA medications received post-RA diag-
nosis, radiographic erosions, and number of samples tested [16].
Serum autoantibody assays
ACPA was determined using a commercially-available second-gen-
eration anti-CCP2 ELISA (Diastat, Axis-Shield Diagnostics, Dundee,
Scotland); CCP2 positivity was based on the manufacturers recom-
mendation at a level of > 5 U/ml. Serum samples also were evaluated for
26 specic ACPAs using a bead-based multiplex antigen array that
measures antibody reactivity to a panel of putative citrullinated auto-
antigens [19]. To reduce the chance of false discovery, analyses of
antigen-specic ACPAs were limited to antibodies targeting citrullinated
forms of vimentin, alpha-enolase, and histone, which are autoantigens
consistently implicated in RA pathogenesis [2022]. IgA rheumatoid
factor (RF), IgG RF, and IgM RF concentrations (IU/ml) were deter-
mined using ELISA (Inova Diagnostics, San Diego, CA). RF positivity was
based on concentrations for each isotype (IgA RF, IgG RF, and IgM RF)
determined to be present in < 2% of controls.
Serum bacterial antibodies
Serum concentrations of IgG antibodies to outer membrane antigens
(OMA) of P gingivalis, P. intermedia, and F. nucleatum were measured by
ELISA, as described in a previous publication from our group [14].
Ethical considerations
The Institutional Review Boards approved the study protocol at the
DoDSR, Walter Reed National Military Medical Center, and the Uni-
versity of Colorado Multiple Institutional Review Board.
Statistical analyses
Participant characteristics were compared between RA and control
groups using chi-square tests, exact chi-square tests, t-tests, or Wilcoxon
rank sum tests as necessary. Autoantibodies and bacterial antibodies
were log (base 2) transformed for all analyses. The primary analysis
investigated the associations between anti-P. gingivalis serum antibodies
and RA diagnosis (i.e., case status). Anti-P. intermedia and anti-
F. nucleatum were evaluated to determine whether associations observed
were specic to P. gingivalis or conversely related to a broader dysbiosis
observed in PD. Initial analyses compared anti-bacterial antibody con-
centrations and biomarkers (i.e., anti-CCP2, ACPA ne specicities
targeting vimentin, histone and alpha-enolase, and RF isotypes) be-
tween groups in the pre-RA diagnosis sample that was closest to diag-
nosis, and the post-RA diagnosis sample using Wilcoxon rank sum tests.
The timing of elevations in anti-bacterial concentrations were evaluated
in RA cases versus controls in a manner previously described [16,18].
Briey, we used mixed models for each bacterial concentration with a
continuous time effect modeled using B-splines and assuming a multi-
variate normal distribution for random subject intercepts and slopes. At
each month prior to diagnosis, we compared autoantibody concentra-
tions to identify the rst instance where concentrations differed signif-
icantly (p <0.05) between cases and controls. These multiple
comparisons were using a stepdown Holm-simulated method. Correla-
tions between anti-bacterial antibody concentrations were evaluated by
Pearson correlation coefcient.
Secondary analyses examined potential associations between anti-
P. gingivalis, anti-P. intermedia, and anti-F. nucleatum antibody concen-
trations and biomarkers (i.e., anti-CCP2, ACPA ne specicities target-
ing vimentin, histone and alpha-enolase, and RF isotypes) within the RA
J.A. Lee et al.
Seminars in Arthritis and Rheumatism 59 (2023) 152176
3
group. These analyses were completed using both unadjusted and
adjusted mixed-effects linear regression models with either RF or ACPA
as the dependent variable, a xed effect for each of the anti-bacterial
antibodies in turn, and random subject intercepts. The adjusted
models also included terms for age, sex, and smoking status.
All analyses were performed utilizing SAS v9.4 (SAS Institute, Cary,
NC).
Results
Participant characteristics and autoantibody values
Patient characteristics and median autoantibody concentrations of
the participants are shown in Table 1. RA cases were slightly more likely
than controls to be ever smokers (32% vs. 23%, p = 0.05); however,
when analysis was limited to non-missing data, RA cases and controls
did not differ with respect to ever smokers (p = 0.15). Higher median
serum concentrations of anti-CCP2, ACPA ne specicities targeting
vimentin, histone, alpha-enolase, and IgA, IgG and IgM RF isotypes were
observed for the immediate/closest pre-diagnosis sample and post-RA
diagnosis sample in RA cases versus controls (p<0.001). Likewise,
anti-CCP2 positivity and IgA, IgG, and IgM RF isotype positivity were
signicantly higher for the immediate/closest pre-diagnosis sample and
post-RA diagnosis sample in RA cases versus controls (p<0.001)
(Table 1).
Serum anti-bacterial antibodies in RA cases versus controls
Median anti-P. gingivalis serum antibody concentrations were not
signicantly different between RA cases and controls with respect to the
immediate/closest pre-diagnosis or post-diagnosis samples (Table 2).
Median anti-P. intermedia serum antibody concentrations were signi-
cantly higher in RA cases than controls for the immediate/closest pre-
diagnosis sample (p = 0.008), but not in the post-diagnosis sample. In
contrast, median anti-F. nucleatum serum antibody concentrations were
lower in RA cases than controls in the immediate/closest pre-diagnosis
sample (p = 0.045) but did not differ in post-diagnosis samples.
Association between pre-RA diagnosis serum anti-bacterial antibody
concentrations and future RA case status
Temporal relationships of anti-P. gingivalis, anti-P. intermedia, and
anti-F. nucleatum serum antibody concentrations with RA cases and
controls are shown in Fig. 1. No evidence of case-control divergence in
anti-P. gingivalis and anti-P. intermedia was observed during the pre-RA
diagnosis period. Anti-F. nucleatum displayed evidence of slight case-
control divergence at 13 years, 7 months prior to diagnosis, with the
controls having higher anti-bacterial antibodies than the cases, but
values reconverged and were not signicantly different at all later time
points. Correlations among the anti-bacterial serum antibody concen-
trations were moderately strong and positive (r = 0.460.66; data not
shown).
Autoantibody concentrations among RA cases and associations with anti-
bacterial antibodies
In analyses limited to RA cases, using data from all pre-diagnosis
observations, anti-P. gingivalis and anti-F. nucleatum serum antibody
concentrations were not signicantly associated with any of the RA
autoantibodies in either unadjusted analyses or in multivariable models
adjusted for age, sex, and smoking status (Table 3).
However, higher anti-P. intermedia serum antibody concentrations
were signicantly associated with higher concentrations of anti-CCP2,
ACPA ne specicities targeting vimentin, histone, alpha-enolase, and
IgA RF autoantibodies (p<0.001) in both unadjusted and adjusted an-
alyses. Anti-P. intermedia serum antibody concentrations were also
Table 1
Patient characteristics and autoantibody values.
Characteristic RA Cases
n = 214
Controls
n = 210
p-value
Age at time of diagnosis, mean (SD) 36.8 (7.9) 36.7 (8.0) 0.89
a1
Sex, n (%)
0.93
a2
Female 102 (48) 101 (48)
Male 112 (52) 109 (52)
Ethnicity, n (%)
b
1.00
a3
White 123 (59) 122 (60)
Black 58 (28) 55 (27)
Hispanic 18 (9) 18 (9)
Asian 5 (2) 5 (2)
American Indian 4 (2) 4 (2)
Other 1 (0) 1 (0)
Ever Smoker, n (%)
c
68 (32) 49 (23) 0.05
a2
Anti-CCP2, U/ml, median (IQ range)
d
Immediate / closest pre-diagnosis
sample
59 (2, 216) 0.3 (0.1, 1.0) <0.001
a4
Post diagnosis sample 52 (3, 204) 0.4 (0.1, 0.9) <0.001
a4
Anti-CCP2, U/ml, n (% positive)
d
Immediate / closest pre-diagnosis
sample
152 (72) 3 (1) <0.001
a3
Post diagnosis sample 153 (72) 0 (0) <0.001
a3
ACPA against vimentin, MFI, median
(IQ range)
d
Immediate / closest pre-diagnosis
sample
288 (93,
1725)
60 (47, 79) <0.001
a4
Post diagnosis sample 397 (86,
1849)
53 (47, 68) <0.001
a4
ACPA against histone, MFI, median
(IQ range)
d
Immediate / closest pre-diagnosis
sample
591 (133,
2563)
91 (71, 126) <0.001
a4
Post diagnosis sample 546 (122,
2478)
77 (62, 108) <0.001
a4
ACPA against alpha-enolase, MFI,
median (IQ range)
d
Immediate / closest pre-diagnosis
sample
310 (108,
4431)
82 (67, 103) <0.001
a4
Post diagnosis sample 406 (112,
3640)
78 (65, 98) <0.001
a4
IgA RF, IU/ml, median (IQ range)
d
Immediate / closest pre-diagnosis
sample
5.8 (2.1,
27.1)
1.3 (0.9, 2.0) <0.001
a4
Post diagnosis sample 5.7 (1.8,
29.3)
1.2 (0.9, 2.0) <0.001
a4
IgA RF, IU/ml, n (% positive)
d
Immediate / closest pre-diagnosis
sample
86 (41) 3 (1) <0.001
a3
Post diagnosis sample 86 (40) 4 (4) <0.001
a3
IgG RF, IU/ml, median (IQ range)
d
Immediate / closest pre-diagnosis
sample
6.4 (4.7,
11.6)
4.5 (3.5, 5.7) <0.001
a4
Post diagnosis sample 6.7 (4.2,
11.7)
4.4 (3.3, 5.8) <0.001
a4
IgG RF, IU/ml, n (% positive)
d
Immediate / closest pre-diagnosis
sample
40 (19) 5 (2) <0.001
a3
Post diagnosis sample 35 (16) 1 (1) <0.001
a3
IgM RF, IU/ml, median (IQ range)
d
Immediate / closest pre-diagnosis
sample
30 (8, 105) 3.8 (2.1, 7.0) <0.001
a4
Post diagnosis sample 30 (8, 105) 3.6 (2.2, 7.7) <0.001
a4
IgM RF, IU/ml, n (% positive)
d
Immediate / closest pre-diagnosis
sample
112 (53) 7 (3) <0.001
a3
Post diagnosis sample 112 (53) 4 (4) <0.001
a3
RA medications (Ever Used), n (%)
Methotrexate 187 (88)
Anti-TNF inhibitor 157 (74)
Radiographic erosions, n (%) 95 (45)
Number of samples tested, per
individual, n (%)
2 0 (0) 1 (0)
3 3 (1) 102 (49)
4 211 (99) 107 (51)
(continued on next page)
J.A. Lee et al.
Seminars in Arthritis and Rheumatism 59 (2023) 152176
4
signicantly associated with IgG RF (p = 0.047, 0.049) and IgM RF (p =
0.003, 0.004) for unadjusted and adjusted values, respectively.
Discussion
This study shows serum anti-P. intermedia antibodies demonstrated
signicant associations with anti-CCP2, ACPA ne specicities targeting
vimentin, histone, alpha-enolase, and IgA, IgG, and IgM RF autoanti-
body concentrations prior to RA diagnosis even after adjusting for age,
sex, and smoking. In contrast, anti-P. gingivalis and anti-F. nucleatum
serum antibody concentrations were not signicantly associated with
RA autoantibodies. Additionally, no longitudinal elevations of anti-
P. gingivalis, anti-P. intermedia, and anti-F. nucleatum serum antibody
concentrations were observed in RA patients prior to a diagnosis of RA
compared to controls.
Prior studies have evaluated serum anti-P. gingivalis antibody con-
centrations in association with pre-RA case status [2325]. Fisher et al.
evaluated a southern European population prior to the onset of RA and
reported the association between smoking and antibodies to P. gingivalis
arginine gingipain (RgpB), and citrullinated PPAD peptides with the risk
of RA and pre-RA autoimmunity [23]. Median timing from blood sam-
pling to diagnosis in pre-RA cases was seven years. Their results showed
that smoking was signicantly associated with an increased risk of RA
before clinical onset of disease and former smoking was associated with
ACPA positivity in pre-RA cases. Antibodies to RgpB and PPAD peptides
were not associated with risk of RA or with pre-RA autoimmunity.
Similar to our study, P. gingivalis antibody was not associated with
pre-RA autoimmunity or risk of RA and the authors suggested this or-
ganism may not play a role in the association between PD and RA in this
cohort [23].
A study by Johansson analyzed a Northern Swedish population and
investigated whether anti-P. gingivalis antibody levels pre-dated the
onset of RA symptoms and ACPA production [24]. The median time
blood samples pre-dated RA symptoms was approximately ve years. In
contrast to the Fisher et al. study [23], their data demonstrated signif-
icantly increased anti-RgpB IgG levels in pre-symptomatic patients and
in RA patients compared with controls. Enhanced levels of antibodies to
a citrullinated PPAD peptide (anti-CPP3) were also found in both
pre-symptomatic and RA individuals. Interestingly, no signicant asso-
ciation was noted between anti-RgpB and anti-CPP3 antibodies. This
study supported a relationship between P. gingivalis and RA by
Table 1 (continued )
Span of pre-RA samples in years,
mean (SD)
5.1 (5.7)
Span, oldest to newest sample, in
years, mean (SD)
e
12.8 (5.2) 12.2 (4.9) 0.30
a1
ACPA = anti-citrullinated protein antibodies.
MFI = mean uorescent intensity.
a1
t-test.
a2
Pearson chi-square test.
a3
Exact Pearson chi-square test.
a4
Wilcoxon
rank-sum test.
b
Each ethnicity group was missing values for 5 cases and 5 controls.
c
Data missing regarding ‘ever smokingin 5 cases and 89 controls (imputed as
never smokers); when analysis limited to non-missing data, ever smoking
observed in 33% of cases and 41% of controls (p = 0.15).
d
Immediate pre-diagnosis samples available for 212 cases and 207 controls;
post-diagnosis samples available for 214 cases and 112 controls.
e
Among 214 cases and 112 controls with a post diagnosis/index date sample.
Table 2
Serum anti-bacterial antibodies in RA cases versus controls.
Serum anti-bacterial antibodies RA Cases
n = 214
Controls
n = 210
p-
value
Anti-P. gingivalis, ug/ml, median (IQ
range)
a
Immediate / closest pre-diagnosis
sample
47 (29, 83) 50 (30, 86) 0.633
Post diagnosis sample 50 (31, 84) 53 (30, 87) 0.913
Anti-P. intermedia, ug/ml, median
(IQ range)
a
Immediate / closest pre-diagnosis
sample
331 (258,
412)
313 (220,
379)
0.008
Post diagnosis sample 372 (289,
445)
371 (289,
423)
0.404
Anti-F. nucleatum, ug/ml, median (IQ
range)
a
Immediate / closest pre-diagnosis
sample
57 (34, 86) 61 (39, 105) 0.045
Post diagnosis sample 57 (33, 96) 67 (42, 97) 0.192
a
Immediate pre-diagnosis samples available for 212 cases and 207 controls;
post-diagnosis samples available for 214 cases and 112 controls.
Fig. 1. Pre-rheumatoid arthritis diagnosis concentrations of serum anti-bacterial antibodies (RA cases shown with solid lines, controls shown with dashed lines).
J.A. Lee et al.
Seminars in Arthritis and Rheumatism 59 (2023) 152176
5
demonstrating increased concentrations of anti-P. gingivalis antibodies in
RA patients compared to controls, detectable years before symptom
development [24].
Manoil et al. measured serum IgG antibodies against selected peri-
odontal pathogens, including P. gingivalis, to determine whether they
were associated with early symptoms or RA development [25]. This
study did not nd an association between serum IgG titers against in-
dividual periodontal pathogens and specic preclinical phases of RA
development. However, the authors found an association between cu-
mulative IgG titers against periodontal pathogens and ACPA-positivity.
These data suggest that synergy among periodontal pathogens, rather
than specic bacterial associations, may be associated with ACPA
development [25].
Our results may differ from prior reports given differences in the
populations studied. In the present study, antibody to P. gingivalis was
directed against outer membrane antigens (OMA), rather than only to
specic P. gingivalis virulence factors seen in the other two studies [23,
24]. Also, the majority of our study population was male and consisted
of active United States military personnel compared to individuals
residing in Northern [24] or Southern Europe [23]. Furthermore, the
mean age of the RA cases in the European studies were around 50 years
old and had a high percentage of ever smokers, ranging from 59% [23]
to 67% [24], while our RA participants averaged 37 years old and had
lower smoking prevalence of 32%. With the differences in age at disease
onset, our younger cohort could suggest a high genetic burden for RA.
That high genetic risk could potentially attenuate the importance of
environmental factors in this population, such as smoking and bacterial
infection leading to PD [26]. We did not determine HLA-SE in the cur-
rent study, although a previous publication by our group found no ev-
idence of an interaction of PD with HLA-DRB1 SE positivity [26].
In our previous study, relationship of P. gingivalis with RA autoan-
tibodies in individuals at high riskfor RA was examined [14]. Patients
were considered autoantibody positive with one or more positive
autoantibody tests and high-risk individuals were either ACPA-positive
or were positive on two or more RF assays. No patients satised the
1987 American College of Rheumatology RA classication criteria [17].
Anti-P. gingivalis concentrations were higher in both the high-risk and
autoantibody positive groups than in the autoantibody negative group.
There were no differences between groups with respect to
anti-P. intermedia or anti-F. nucleatum. The majority of this cohort was
slightly older and predominantly female when compared to our
younger, male population and could account for the different associa-
tions with serum anti-bacterial antibodies [14]. These contrasting con-
clusions suggest additional research is needed to further explore
whether antibody concentrations to the pathogen P. gingivalis may be
increased prior to onset of RA symptoms and linked to the development
of RA.
Although P. gingivalis is the most studied periodontal microorganism
in the pathogenesis of RA, it has been suggested that P. intermedia may
also play a role in RA progression, albeit by a different mechanism. A
study by Schwenzer et al. suggested that, since P. intermedia does not
express a PAD, its ability to induce ACPA differs from P. gingivalis [12]
potentially through a mechanism whereby degradation of neutrophil
extracellular traps (NETs) by nucleases from P. intermedia leads to the
release of PADs [27] and increases the pathogenicity of this organism
[28]. Kimura et al. [29] evaluated synovitis and its association with
periodontal pathogens and established biomarkers of RA. Greater
P. intermedia antibody titer was observed in active RA patients and RA
patients in clinical remission with subclinical synovitis, detected by ul-
trasound, compared to RA patients in clinical remission without sub-
clinical synovitis. An association of P. intermedia antibody titer and
disease activity of RA, specically synovitis, was proposed. The mech-
anism suggested by the authors is activation of macrophages by
P. intermedia which initiates production of IL-6 and TNF-
α
, inammatory
cytokines that play a role in periodontal and joint destruction. Of note,
Scher et al. reported that Prevotella and Leptotrichia species were the only
characteristic taxa in the oral microbiota in the new-onset RA group
irrespective of PD status and were completely absent in the oral
microbiota of controls [30]. While other investigators were unable to
demonstrate a relationship between P. intermedia and RA [31,32], our
study observed a strong association with anti-CCP2, certain ACPA
specicities as well as several isotypes of RF and highlights a need to
further explore the potential role of P. intermedia in RA pathogenesis
and, in particular, the generation of these RA-related autoantibodies.
Limited studies exist evaluating anti-F. nucleatum antibody concen-
trations with RA. One study analyzed saliva samples of early RA patients
and found microbiota rich in F. nucleatum when compared to healthy
controls and proposed the oral microbiota may be useful in detecting
Table 3
Associations of all pre-diagnosis autoantibody sample concentrations among RA
cases with serum anti-bacterial antibodies.
Unadjusted Adjusted
a
Dependent
Variable
Anti-P. gingivalis
coefcient (95%
CI)
p-value Anti-P. gingivalis
coefcient (95%
CI)
p-value
Anti-CCP2 0.101 ( 0.274,
0.475)
0.597 0.105 ( 0.274,
0.484)
0.586
ACPA against
vimentin
0.020 ( 0.180,
0.220)
0.845 0.032 ( 0.170,
0.233)
0.758
ACPA against
histone
0.168 ( 0.027,
0.363)
0.092 0.173 ( 0.024,
0.371)
0.085
ACPA against
alpha-
enolase
0.073 ( 0.132,
0.277)
0.485 0.094 ( 0.111,
0.299)
0.367
IgA RF 0.066 ( 0.244,
0.112)
0.465 0.060 ( 0.239,
0.119)
0.510
IgG RF 0.038 ( 0.057,
0.133)
0.434 0.040 ( 0.056,
0.137)
0.411
IgM RF 0.019 ( 0.158,
0.195)
0.835 0.030 ( 0.144,
0.205)
0.733
Dependent
Variable
Anti-P. intermedia
coefcient (95%
CI)
p-
value
Anti-P. intermedia
coefcient (95%
CI)
p-
value
Anti-CCP2 1.838 (1.210,
2.466)
<0.001 1.869 (1.235,
2.503)
<0.001
ACPA against
vimentin
0.792 (0.457,
1.127)
<0.001 0.827 (0.490,
1.163)
<0.001
ACPA against
histone
0.739 (0.410,
1.068)
<0.001 0.758 (0.426,
1.090)
<0.001
ACPA against
alpha-
enolase
0.790 (0.443,
1.136)
<0.001 0.840 (0.492,
1.187)
<0.001
IgA RF 0.525 (0.236,
0.814)
<0.001 0.536 (0.245,
0.827)
<0.001
IgG RF 0.163 (0.002,
0.325)
0.047 0.163 (0.001,
0.326)
0.049
IgM RF 0.454 (0.157,
0.750)
0.003 0.442 (0.146,
0.737)
0.004
Dependent
Variable
Anti-F. nucleatum
coefcient (95%
CI)
p-
value
Anti-F. nucleatum
coefcient (95%
CI)
p-
value
Anti-CCP2 0.066 ( 0.326,
0.459)
0.740 0.081 ( 0.314,
0.476)
0.687
ACPA against
vimentin
0.055 ( 0.154,
0.264)
0.604 0.048 ( 0.161,
0.256)
0.655
ACPA against
histone
0.036 ( 0.169,
0.241)
0.731 0.041 ( 0.165,
0.247)
0.693
ACPA against
alpha-
enolase
0.179 ( 0.034,
0.392)
0.100 0.181 ( 0.032,
0.393)
0.095
IgA RF 0.090 ( 0.275,
0.094)
0.337 0.081 ( 0.266,
0.104)
0.388
IgG RF 0.080 ( 0.020,
0.179)
0.117 0.084 ( 0.016,
0.184)
0.098
IgM RF 0.046 ( 0.231,
0.138)
0.622 0.024 ( 0.206,
0.158)
0.799
ACPA = anti-citrullinated protein antibodies.
All measures in this table were log base 2 transformed.
a
Models were adjusted for age, sex, and smoking.
J.A. Lee et al.
Seminars in Arthritis and Rheumatism 59 (2023) 152176
6
risk assessment for early onset of RA [13]. In looking at subgingival
biolm of RA patients, F. nucleatum was found in higher concentrations
in aCCP-positive patients with RA versus controls, though this nding
was not statistically signicant [33]. In a separate study, F. nucleatum
was found in the synovial uid of RA patients derived from both native
and prosthetic joints. Identical clones of the bacteria were found in the
same patients plaque sample, and it was proposed that F. nucleatum can
translocate from the oral cavity to the synovial cavity [34]. In contrast,
our data does not provide compelling evidence to support a role of
F. nucleatum in RA development. In contrast to prior reports, our results
demonstrated only a slight case-control divergence of anti-F. nucleatum
prior to RA diagnosis; however, controls had initially higher concen-
trations that reconverged to no longer be statistically signicant than
concentrations in RA cases. When compared to P. gingivalis and P.
intermedia, potential mechanisms linking F. nucleatum and RA risk
remain poorly understood.
There are limitations in this study. The participants were military
personnel with a relatively high proportion of men to women (52% vs.
48%, respectively) and a younger age of RA onset (37 years old). Thus,
these results may not be generalizable to other RA populations [35]. A
majority of RA cases utilized methotrexate and/or biologics (88% and
74%, respectively), which could have impacted these results. Further-
more, there were only 112 control patient samples available for post-RA
diagnosis evaluations. The lack of a difference in anti-P. intermedia
concentrations in RA cases versus controls post-RA diagnosis needs to be
interpreted with caution in light of this smaller sample size available for
analysis. In addition, most of the pre-RA serum samples were collected
within 5 years of diagnosis, which could have limited our ability to
detect earlier differences in anti-bacterial or autoantibody elevations
[18]. In future studies, more frequent serum sample collection over
more extended time periods would provide an even more comprehen-
sive look at the autoantibody and anti-bacterial responses potentially
leading to RA onset. PD status was not determined in this study and,
therefore, we were unable to associate periodontal status with the pa-
tientssystemic response against the periodontal pathogens investi-
gated. Moreover, the taxa could exert a local response without triggering
a serum IgG response; therefore, null associations should be carefully
considered. Finally, future studies also should focus on the plethora of
inammatory reactions occurring in the gingival tissues that have the
potential to stimulate autoantibody production associated with RA.
Conclusion
In conclusion, no longitudinal elevations of serum anti-bacterial
antibody concentrations were observed in RA patients prior to a diag-
nosis of RA compared to controls. However, anti-P. intermedia displayed
a signicant association with RA autoantibody concentrations prior to
RA diagnosis, suggesting a potential role of this organism in progression
towards clinically-detectable RA.
Funding statement
Dr. Deane receives research support from the Rheumatology
Research Foundation, Department of Defense Congressionally Directed
Medical Research Program grants PR120839 and PR191079, NIH/
NCATS Colorado CTSA Grant Number UL1 TR001082 and NIH/NIAMS
P30 AR079369.
Dr. Mikuls receives research support from the VA (Merit grant
BX004600), the US Department of Defense (PR200793), and NIH/
NIGMS (U54GM115458).
Disclosure statement
Dr. Deane has received kits for testing RA-related autoantibodies
from Werfen through research relationships. There are no other nan-
cial disclosures for the other authors.
Military disclaimer
The identication of specic products or scientic instrumentation is
considered an integral part of the scientic endeavor and does not
constitute endorsement or implied endorsement on the part of the
author(s), DoD, or any component agency. The views expressed in this
article are those of the author(s) and do not necessarily reect the
ofcial policy of the Department of Defense or the U.S. Government.
CRediT authorship contribution statement
Joyce A. Lee: Conceptualization, Investigation, Methodology,
Visualization, Writing original draft, Writing review & editing. Ted
R. Mikuls: Conceptualization, Funding acquisition, Investigation,
Methodology, Project administration, Supervision, Visualization,
Writing original draft, Writing review & editing. Kevin D. Deane:
Conceptualization, Data curation, Funding acquisition, Investigation,
Project administration, Resources, Writing review & editing. Harlan
R. Sayles: Formal analysis, Validation, Visualization, Writing original
draft, Writing review & editing. Geoffrey M. Thiele: Data curation,
Investigation, Methodology, Resources, Validation, Writing review &
editing. Jess D. Edison: Data curation, Investigation, Project adminis-
tration, Supervision, Writing review & editing. Brandie D. Wagner:
Methodology, Software, Writing review & editing. Marie L. Feser:
Project administration, Resources, Writing review & editing. Laura K.
Moss: Data curation, Writing review & editing. Lindsay B. Kelmen-
son: Data curation, Writing review & editing. William H. Robinson:
Data curation, Investigation, Project administration, Resources, Vali-
dation, Writing review & editing. Jeffrey B. Payne: Conceptualiza-
tion, Investigation, Methodology, Project administration, Supervision,
Visualization, Writing original draft, Writing review & editing.
Data Availability
Data requests can be made to the authors although use is restricted
based on Department of Defense guidelines.
References
[1] Mikuls TR, Payne JB, Deane KD, Thiele GM. Autoimmunity of the lung and oral
mucosa in a multisystem inammatory disease: the spark that lights the re in
rheumatoid arthritis? J Allergy Clin Immunol 2016;137(1):2834. https://doi.org/
10.1016/j.jaci.2015.10.024. PubMed PMID: 26768760.
[2] Loos BG, Van Dyke TE. The role of inammation and genetics in periodontal
disease. Periodontol 2000 2020;83(1):2639. https://doi.org/10.1111/prd.12297.
PubMed PMID: 32385877; PubMed Central PMCID: PMC7319430.
[3] Bartold PM, Lopez-Oliva I. Periodontitis and rheumatoid arthritis: an update 2012-
2017. Periodontol 2000 2020;83(1):189212. https://doi.org/10.1111/prd.12300.
PubMed PMID: 32385878.
[4] Unriza-Puin S, Bautista-Molano W, Lafaurie GI, Valle-Onate R, Chalem P, Chila-
Moreno L, et al. Are obesity, ACPAs and periodontitis conditions that inuence the
risk of developing rheumatoid arthritis in rst-degree relatives? Clin Rheumatol
2017;36(4):799806. https://doi.org/10.1007/s10067-016-3519-z. Epub
20161228PubMed PMID: 28028684.
[5] Lee KH, Choi YY. Rheumatoid arthritis and periodontitis in adults: using the
Korean National Health Insurance Service-National Sample Cohort. J Periodontol
2020;91(9):118693. https://doi.org/10.1002/JPER.19-0311. Epub
20200214PubMed PMID: 31984496.
[6] Qiao Y, Wang Z, Li Y, Han Y, Zhou Y, Cao X. Rheumatoid arthritis risk in
periodontitis patients: a systematic review and meta-analysis. Joint Bone Spine
2020;87(6):55664. https://doi.org/10.1016/j.jbspin.2020.04.024. Epub
20200625PubMed PMID: 32593704.
[7] Scher JU, Bretz WA, Abramson SB. Periodontal disease and subgingival microbiota
as contributors for rheumatoid arthritis pathogenesis: modiable risk factors? Curr
Opin Rheumatol 2014;26(4):4249. https://doi.org/10.1097/
BOR.0000000000000076. PubMed PMID: 24807405; PubMed Central PMCID:
PMC4128331.
[8] How KY, Song KP, Chan KG. Porphyromonas gingivalis: an overview of
periodontopathic pathogen below the gum line. Front Microbiol 2016;7:53.
https://doi.org/10.3389/fmicb.2016.00053. Epub 20160209PubMed PMID:
26903954; PubMed Central PMCID: PMC4746253.
[9] Hajishengallis G, Darveau RP, Curtis MA. The keystone-pathogen hypothesis. Nat
Rev Microbiol 2012;10(10):71725. https://doi.org/10.1038/nrmicro2873. Epub
20120903PubMed PMID: 22941505; PubMed Central PMCID: PMC3498498.
J.A. Lee et al.
Seminars in Arthritis and Rheumatism 59 (2023) 152176
7
[10] Mikuls TR, Walker C, Qiu F, Yu F, Thiele GM, Alfant B, et al. The subgingival
microbiome in patients with established rheumatoid arthritis. Rheumatology
(Oxford) 2018;57(7):116272. https://doi.org/10.1093/rheumatology/key052.
PubMed PMID: 29562298.
[11] Rosenstein ED, Greenwald RA, Kushner LJ, Weissmann G. Hypothesis: the humoral
immune response to oral bacteria provides a stimulus for the development of
rheumatoid arthritis. Inammation 2004;28(6):3118. https://doi.org/10.1007/
s10753-004-6641-z. PubMed PMID: 16245073.
[12] Schwenzer A, Quirke AM, Marzeda AM, Wong A, Montgomery AB, Sayles HR, et al.
Association of distinct ne specicities of anti-citrullinated peptide antibodies with
elevated immune responses to prevotella intermedia in a subgroup of patients with
rheumatoid arthritis and periodontitis. Arthritis Rheumatol 2017;69(12):230313.
https://doi.org/10.1002/art.40227. Epub 20171030PubMed PMID: 29084415;
PubMed Central PMCID: PMC5711558.
[13] Esberg A, Johansson L, Johansson I, Dahlqvist SR. Oral microbiota identies
patients in early onset rheumatoid arthritis. Microorganisms 2021;9(8). https://
doi.org/10.3390/microorganisms9081657. Epub 20210803PubMed PMID:
34442739; PubMed Central PMCID: PMC8400434.
[14] Mikuls TR, Thiele GM, Deane KD, Payne JB, ODell JR, Yu F, et al. Porphyromonas
gingivalis and disease-related autoantibodies in individuals at increased risk of
rheumatoid arthritis. Arthritis Rheum 2012;64(11):352230. https://doi.org/
10.1002/art.34595. PubMed PMID: 22736291; PubMed Central PMCID:
PMC3467347.
[15] Perdue CL, Eick-Cost AA, Rubertone MV. A brief description of the operation of the
DoD serum repository. Mil Med 2015;180(10 Suppl):102. https://doi.org/
10.7205/MILMED-D-14-00739. PubMed PMID: 26444888.
[16] Kelmenson LB, Wagner BD, McNair BK, Frazer-Abel A, Demoruelle MK,
Bergstedt DT, et al. Timing of elevations of autoantibody isotypes prior to diagnosis
of rheumatoid arthritis. Arthritis Rheumatol 2020;72(2):25161. https://doi.org/
10.1002/art.41091. Epub 20191226PubMed PMID: 31464042; PubMed Central
PMCID: PMC6994340.
[17] Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF, Cooper NS, et al. The
American Rheumatism Association 1987 revised criteria for the classication of
rheumatoid arthritis. Arthritis Rheum 1988;31(3):31524. https://doi.org/
10.1002/art.1780310302. PubMed PMID: 3358796.
[18] Mikuls TR, Edison J, Meeshaw E, Sayles H, England BR, Duryee MJ, et al.
Autoantibodies to malondialdehyde-acetaldehyde are detected prior to rheumatoid
arthritis diagnosis and after other disease specic autoantibodies. Arthritis
Rheumatol 2020;72(12):20259. https://doi.org/10.1002/art.41424. Epub
20201015PubMed PMID: 32621635; PubMed Central PMCID: PMC7722146.
[19] Gonzalez SM, Payne JB, Yu F, Thiele GM, Erickson AR, Johnson PG, et al. Alveolar
bone loss is associated with circulating anti-citrullinated protein antibody (ACPA)
in patients with rheumatoid arthritis. J Periodontol 2015;86(2):22231. https://
doi.org/10.1902/jop.2014.140425. Epub 20141009PubMed PMID: 25299390.
[20] Van Steendam K, Tilleman K, Deforce D. The relevance of citrullinated vimentin in
the production of antibodies against citrullinated proteins and the pathogenesis of
rheumatoid arthritis. Rheumatology (Oxford) 2011;50(5):8307. https://doi.org/
10.1093/rheumatology/keq419. Epub 20110127PubMed PMID: 21278075;
PubMed Central PMCID: PMC3077912.
[21] Lundberg K, Wegner N, Yucel-Lindberg T, Venables PJ. Periodontitis in RA-the
citrullinated enolase connection. Nat Rev Rheumatol 2010;6(12):72730. https://
doi.org/10.1038/nrrheum.2010.139. Epub 20100907PubMed PMID: 20820197.
[22] Mergaert AM, Bawadekar M, Nguyen TQ, Massarenti L, Holmes CL, Rebernick R,
et al. Reduced anti-histone antibodies and increased risk of rheumatoid arthritis
associated with a single nucleotide polymorphism in PADI4 in North Americans.
Int J Mol Sci 2019;20(12). https://doi.org/10.3390/ijms20123093. Epub
20190625PubMed PMID: 31242568; PubMed Central PMCID: PMC6627847.
[23] Fisher BA, Cartwright AJ, Quirke AM, de Pablo P, Romaguera D, Panico S, et al.
Smoking, Porphyromonas gingivalis and the immune response to citrullinated
autoantigens before the clinical onset of rheumatoid arthritis in a Southern
European nested case-control study. BMC Musculoskelet Disord 2015;16:331.
https://doi.org/10.1186/s12891-015-0792-y. Epub 20151104PubMed PMID:
26537917; PubMed Central PMCID: PMC4634856.
[24] Johansson L, Sherina N, Kharlamova N, Potempa B, Larsson B, Israelsson L, et al.
Concentration of antibodies against Porphyromonas gingivalis is increased before
the onset of symptoms of rheumatoid arthritis. Arthritis Res Ther 2016;18:201.
https://doi.org/10.1186/s13075-016-1100-4. Epub 20160907PubMed PMID:
27605245; PubMed Central PMCID: PMC5015325.
[25] Manoil D, Courvoisier DS, Gilbert B, Moller B, Walker UA, Muehlenen IV, et al.
Associations between serum antibodies to periodontal pathogens and preclinical
phases of rheumatoid arthritis. Rheumatology (Oxford) 2021;60(10):475564.
https://doi.org/10.1093/rheumatology/keab097. PubMed PMID: 33512428.
[26] Mikuls TR, Payne JB, Yu F, Thiele GM, Reynolds RJ, Cannon GW, et al.
Periodontitis and Porphyromonas gingivalis in patients with rheumatoid arthritis.
Arthritis Rheumatol 2014;66(5):1090100. https://doi.org/10.1002/art.38348.
PubMed PMID: 24782175; PubMed Central PMCID: PMC4115329.
[27] Spengler J, Lugonja B, Ytterberg AJ, Zubarev RA, Creese AJ, Pearson MJ, et al.
Release of active peptidyl arginine deiminases by neutrophils can explain
production of extracellular citrullinated autoantigens in rheumatoid arthritis
synovial uid. Arthritis Rheumatol 2015;67(12):313545. https://doi.org/
10.1002/art.39313. PubMed PMID: 26245941; PubMed Central PMCID:
PMC4832324.
[28] Doke M, Fukamachi H, Morisaki H, Arimoto T, Kataoka H, Kuwata H. Nucleases
from Prevotella intermedia can degrade neutrophil extracellular traps. Mol Oral
Microbiol 2017;32(4):288300. https://doi.org/10.1111/omi.12171. Epub
20160920PubMed PMID: 27476978; PubMed Central PMCID: PMC5516193.
[29] Kimura Y, Yoshida S, Takeuchi T, Kimura M, Yoshikawa A, Hiramatsu Y, et al.
Periodontal pathogens participate in synovitis in patients with rheumatoid arthritis
in clinical remission: a retrospective case-control study. Rheumatology (Oxford)
2015;54(12):225763. https://doi.org/10.1093/rheumatology/kev274. Epub
20150805PubMed PMID: 26248812.
[30] Scher JU, Ubeda C, Equinda M, Khanin R, Buischi Y, Viale A, et al. Periodontal
disease and the oral microbiota in new-onset rheumatoid arthritis. Arthritis Rheum
2012;64(10):308394. https://doi.org/10.1002/art.34539. PubMed PMID:
22576262; PubMed Central PMCID: PMC3428472.
[31] Martinez-Rivera JI, Xibille-Friedmann DX, Gonzalez-Christen J, de la Garza-
Ramos MA, Carrillo-Vazquez SM, Montiel-Hernandez JL. Salivary ammonia levels
and Tannerella forsythia are associated with rheumatoid arthritis: a cross sectional
study. Clin Exp Dent Res 2017;3(3):10714. https://doi.org/10.1002/cre2.68.
Epub 20170607PubMed PMID: 29744187; PubMed Central PMCID: PMC5719825.
[32] Sato K, Takahashi N, Kato T, Matsuda Y, Yokoji M, Yamada M, et al. Aggravation of
collagen-induced arthritis by orally administered Porphyromonas gingivalis
through modulation of the gut microbiota and gut immune system. Sci Rep 2017;7
(1):6955. https://doi.org/10.1038/s41598-017-07196-7. Epub 20170731PubMed
PMID: 28761156; PubMed Central PMCID: PMC5537233.
[33] Schmickler J, Rupprecht A, Patschan S, Patschan D, Muller GA, Haak R, et al.
Cross-sectional evaluation of periodontal status and microbiologic and rheumatoid
parameters in a large cohort of patients with rheumatoid arthritis. J Periodontol
2017;88(4):36879. https://doi.org/10.1902/jop.2016.160355. Epub
20161118PubMed PMID: 27858553.
[34] Temoin S, Chakaki A, Askari A, El-Halaby A, Fitzgerald S, Marcus RE, et al.
Identication of oral bacterial DNA in synovial uid of patients with arthritis with
native and failed prosthetic joints. J Clin Rheumatol 2012;18(3):11721. https://
doi.org/10.1097/RHU.0b013e3182500c95. PubMed PMID: 22426587; PubMed
Central PMCID: PMC3888235.
[35] Gabriel SE, Crowson CS, OFallon WM. The epidemiology of rheumatoid arthritis in
Rochester, Minnesota, 1955-1985. Arthritis Rheum 1999;42(3):41520. https://
doi.org/10.1002/1529-0131(199904)42:3<415::AID-ANR4>3.0.CO;2-Z. PubMed
PMID: 10088762.
J.A. Lee et al.