Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 May 30.
Published in final edited form as: Rheum Dis Clin North Am. 2012 May 30;38(2):405–426. doi: 10.1016/j.rdc.2012.04.002

Environmental and gene-environment interactions and risk of rheumatoid arthritis

Elizabeth W Karlson 1, Kevin Deane 2
PMCID: PMC3402910  NIHMSID: NIHMS381766  PMID: 22819092

Abstract

Multiple environmental factors including hormones, dietary factors, infections and exposure to tobacco smoke as well as gene-environment interactions have been associated with increased risk for rheumatoid arthritis (RA). Importantly, the growing understanding of the prolonged period prior to the first onset of symptoms of RA suggests that these environmental and genetic factors are likely acting to drive the development of RA-related autoimmunity long before the appearance of the first joint symptoms and clinical findings that are characteristic of RA. Herein we will review these factors and interactions, especially those that have been investigated in a prospective fashion prior to the symptomatic onset of RA. We will also discuss how these factors may be explored in future study to further the understanding of the pathogenesis of RA, and ultimately perhaps develop preventive measures for this disease.

Keywords: rheumatoid arthritis, environmental risk factors, gene-environment interactions

Introduction

While etiology of rheumatoid arthritis (RA) is unknown, a growing body of evidence suggests that it develops in individuals with inherited genetic risk factors after exposure to environmental triggers. The identification of autoantibodies and cytokines in the serum many years prior to the diagnosis of RA led to conceptualization of the development of RA as occurring in phases (Figure 1).18 In this model of RA development there is an asymptomatic period of genetic risk in which environmental exposures are encountered, followed by an asymptomatic immune activation phase in which autoantibodies and inflammatory markers are found, likely followed by a phase of articular symptoms in absence of clearly definable arthritis, which is finally followed by the phase with signs of inflammatory arthritis (IA) that is perhaps initially unclassifiable but over time evolves to the point at which it is classifiable as RA by established criteria such as the 1987 American College of Rheumatology (ACR) criteria or the 2010 ACR/European League Against Rheumatism (EULAR) criteria.9,10 Similar phases of development have been proposed in other autoimmune diseases such as type 1 diabetes and systemic lupus erythematosus (SLE).11,12 This suggests a complex series of events in which a genetically susceptible host is exposed to environmental risk factors that trigger autoimmunity with autoantibody production, and a second or more event(s) or exposure(s) that might drive further immune dysregulation and eventual development of symptomatic inflammatory arthritis (IA). The interaction between genetic and environmental factors such as cigarette smoking within the subtype of ACPA positive RA13 provides clues to disease pathogenesis, but much of the complexity of RA etiopathogenesis has yet to be delineated.

Figure 1. Phases in the Development of Rheumatoid Arthritis.

Figure 1

Phase 1) “asymptomatic” phase of genetic risk; Phase 2) asymptomatic “immune activation” phase with autoantibodies and elevated biomarkers such as cytokines and chemokines; Phase 3) “pre-clinical” phase with abnormal biomarkers of inflammation and perhaps arthralgias; and Phase 4) clinically-apparent inflammatory arthritis (defined as symptomatic joint disease including pain, stiffness and swelling, and identifiable synovitis on examination) that may be undifferentiated arthritis (UA), or fulfill classification for RA. Identifying the environmental factors associated with transitions between phases, key exposure windows leading to transition between phases, and the biologic mechanisms involved in these transitions is a challenge for the field.

Epidemiologic research has produced convincing evidence for strong environmental risk factors for RA, including cigarette smoking1427, exogenous hormone use2836 and female reproductive factors.28,3741 Other exposures have also been identified as risk factors for RA including silica4246, air pollution47 and periodontitis48, and some exposures such as alcohol have been reported as protective factors49,50 although these studies have not been widely replicated. Other factors are also associated with increased risk for RA including higher birthweight.51,52 Furthermore, having a first degree relative (FDR) with RA increases RA risk 3- to 9-fold compared to that in the general population suggesting the influence of shared genetic and/or environmental factors.53 Recent whole genome association studies in RA have identified > 30 novel risk loci confirmed by meta-analysis in addition to well established associations with the HLA-DRB1 “shared epitope” (HLA-SE) alleles.54 Gene-environment interactions between HLA-SE and potentially other risk alleles may contribute to RA pathogenesis.5,13,5557 Importantly, models that incorporate genetic, biomarker and lifestyle/environmental factors can now predict the risk of developing RA with greater precision than any of these factors alone.56,58 This review will focus on validated environmental, lifestyle, and dietary exposures in the pre- RA phases prior to the objective signs of IA.

Defining the phases of development of autoimmunity, inflammatory arthritis and early rheumatoid arthritis

Multiple studies have shown that RA-related autoantibodies may be elevated up to years prior to the onset of clinically-apparent IA. Some of these studies were performed in a prospective fashion, with careful joint evaluations in currently asymptomatic individuals.5961 However, most of these studies are somewhat limited in their conclusions about the precise timing of elevations of RA-related autoantibodies before the onset of clinically-apparent IA because they were not performed as part of rigorous prospective evaluations of individuals at-risk for future RA.28,59,6266 Regardless, in sum these studies strongly suggest that RA develops in four distinct phases: 1) an “asymptomatic” phase of genetic risk; 2) an asymptomatic “immune activation” phase in which abnormalities of autoantibodies and biomarkers such as cytokines and chemokines are documented up to 14 years prior to RA15, 7,8,66,67; 3) a “pre-clinical” phase with abnormal biomarkers of inflammation and perhaps arthralgias68,69; and 4) clinically-apparent IA (symptomatic joint disease including pain, stiffness and swelling, and identifiable synovitis on examination) that may be undifferentiated arthritis (UA), or fulfill classification for RA (Figure 1).70,71 Identifying the environmental factors associated with transitions between phases, key exposure windows leading to transition between phases, and the biologic mechanisms involved in these transitions is a challenge for the field.

Epidemiologic study design

Studies of the environmental factors associated with RA are often conducted through a case control design in which cases with RA and healthy controls are surveyed retrospectively for environmental, lifestyle, and behavioral factors occurring prior to disease onset in cases and prior to a matched date (or age) for controls. This study design can be limited by recall bias, where cases recall and report exposures differentially than do controls.72 Also, in case-control studies, results are reported as odds ratios which can approximate relative risk in settings where the sample size is high, but do not provide estimates of population risk. The prospective cohort design is considered less biased, as non-diseased subjects can be followed prospectively exposure assessment while asymptomatic prior to the development of RA. Examples of prospective cohort studies of RA include the US Nurses’ Health Study21, Iowa Women’s Health Study24,73, Malmo Diet and Cancer Study39,41, and more recently Lifelines in the Netherlands. In the Norfolk Arthritis Register (NOAR, Great Britain) cohort study, risk factors for the development of Inflammatory Arthritis (IA) and RA that meets ACR criteria have been studied.74 The advantage of prospective designs is analyses that produce relative risks, the ability to estimate population attributable risks, the ability assess repeated time-varying exposures and to study exposure windows prior to disease onset.

An alternative approach to understanding both timing of environmental exposures and their influence at points of transition in the phases of RA is prospective cohort studies of high risk individuals followed for the development of autoantibodies and immune markers. Two groups have led the field in studies of high risk first degree relatives (FDR): 1) the Studies of the Etiology of RA (SERA) in the United States75 and 2) studies of North American Natives (NAN) in central Canada.76 In each of these studies, FDRs are followed annually for environment, lifestyle and behavioral risk factors and the development of RA-related autoantibodies (RF and ACPA) and immune markers, as well as signs and symptoms of inflammatory arthritis (Table 1). Similar studies of FDRs are being launched in Europe.

Table 1.

Environmental, behavioral and lifestyle risk factors for rheumatoid arthritis, inflammatory arthritis, or autoimmunity in prospective cohort studies

Site Population Cohort Smoking Silica Air
pollution
Reproductive
factors
Birth
Weight
Alcohol Diet Periodontal
Disease
Boston Nurses NHS X X X X X X X*

Iowa Women’s
Health Study
Women IWHS X X X
Sweden General Malmo Diet and
Cancer Study
X X X
Netherlands General Lifelines X X X X X X X**
United Kingdom General NOAR X X X
Colorado FDR SERA X X X X X X*
Manitoba FDR NAN X X X X X**
Geneva FDR Geneva X X X X X X*

X – Exposure ascertained by survey

*

Subjects complete survey regarding periodontitis symptoms

**

Subjects undergo dental examination for periodontitis

Cigarette Smoking Exposure

Modifiable environmental factors contribute substantially to population risk of RA. Multiple case-control and prospective cohort studies have demonstrated that cigarette smoking is the strongest environmental factor linked with RA,14,15,1722,2427,77 and its population attributable risk is 25% for all RA and 35% for seropositive rheumatoid factor(RF+) and anti-citrullinated protein antibody positive (ACPA+) RA.23,27 Relative risks for cigarette smoking range from 1.6 for risk of all RA to 1.8 for risk of seropositive RA.27 The association appears to be stronger for men than for women.17,18,22,25,78 Furthermore, several studies demonstrate a dose-response between heavier smoking and RA, particularly seropositive RA with persistence of RA risk for 20 years after smoking cessation.13,21,23,26,27,77 The specific mechanisms by-which smoking may be related to the generation of RA-related autoimmunity are unknown; however, there are several clues to pathogenesis. Smoking was shown to be associated with an increased levels of citrullinated proteins as well as expression of peptidyl arginine deiminase 2 (PADI2) in pulmonary cells obtained by bronchoalveolar lavage.79 In addition, the risk of seropositive RA associated with smoking has been reported to be highest and in those who carry the HLA-DRB1 shared epitope (HLA-SE).13,77 Recent observations indicate that certain DRB1 alleles, in particular 0401 and 0404, are associated with a strong immunity to citrullinated peptides, such as vimentin or enolase peptides.57,8082 The specific time point in the development of RA that smoking acts to increase risk is unknown; however, in the prospective SERA project, smoking was associated with RF positivity in subjects enriched with HLA-SE, but no RA symptoms, suggesting that smoking may act early in immune dysregulation and may lead to future RA.83 Smoking has been also been associated with more generalized immune abnormalities including alterations in T cell function84,85, reduction in NK cells86, impairment of humoral immunity86,87 and elevated levels of inflammatory markers such as IL-6 and C-reactive protein88,89 as well as elevations of rheumatoid factor (RF) in absence of RA90. While some of these effects associated with smoking such as impaired humoral immunity are somewhat paradoxical when trying to understand the relationship between smoking and development of RA, especially seropositive RA, in sum, these findings suggest that smoking may lead to auto-antigen generation and alteration of other inflammatory factors that in a permissive genetic background leads to the development of RA-related autoimmunity. However, the specific mechanistic details of how smoking triggers RA, including what anatomic site smoking may drive RA-related autoimmunity need further exploration, especially in prospective studies of preclinical RA. Furthermore, recent data suggesting decreased responsiveness to therapy in patients with established RA who are smokers suggests some relationship between disease development and smoking even after the onset of symptomatic IA, although the specific mechanisms of these relationships also need further exploration.91,92

Occupational Exposures

A case control study from Sweden, the Epidemiologic Investigation of RA (EIRA) reported an association between occupational silica exposure and RA among 276 male RA cases and 276 healthy controls with evidence for silica* smoking interaction.42 Silica dust exposure was a risk factor only for RF+ and ACPA+ RA and not for seronegative RA. Exposure to silica dust though the respiratory tract in occupations such as rock drilling, mining and sand blasting, has been linked to RA risk in other epidemiologic studies.4346 High-level exposure to silica dust can cause chronic inflammation and fibrosis in the lung and other organs, and it may be that such inflammation leads to humoral immune responses and hence greater risk for seropositive RA. Furthermore, occupational exposure to other factors such as mineral oils (exposure pathways through the lung and skin) was also investigated in EIRA93 and exposure was associated with increased risk of RF+/ACPA+ RA, and not of seronegative RA. Mineral oil can also act as an adjuvant capable of inducing experimental arthritis in rodent models and may have a similar action in humans.94,95 Substantial exposure to inhaled organic solvents, in occupations such as upholstering, hair-dressing and concrete work, was associated with an increased risk of RA.96 As with smoking, the specific details of how occupational exposures lead to the generation of RA-related autoimmunity, especially seropositive disease, need further study.

Reproductive and Hormonal Factors

Abundant epidemiological, clinical, and experimental evidence implicates hormones in the incidence and clinical expression of RA. Women are 2 to 4 times more likely than men to develop RA.97,98 In women, RA frequently develops at times when sex steroid hormone levels are in flux, such as in the post-partum and perimenopausal periods.99102 In most2835,103, but not all studies16,38,39,104107, oral contraceptive use is protective against the development of RA. Studies such as the NHS demonstrated a strong trend of decreasing risk of RA with increasing total duration of breast feeding38 with inverse findings for breast feeding and RA confirmed in 2 studies39,108 while another study showed a positive association.40 In the NHS irregular menstrual cycles and age at menarche ≤ 10 were associated with increased RA risk38; another study demonstrated that age at menarche ≤ 12 and younger age at menopause were inversely associated with RA risk.41 Studies suggest that RA incidence rises with age97,109 with a peak incidence at menopause.38,99,110 However, exogenous estrogen therapy among post-menopausal women did not reduce RA risk.36,38 In the SERA cohort, among subjects enriched with HLA-SE alleles, oral contraceptive pill use was associated with decreased risk for RF positivity, independent of age, education, and smoking. This finding may indicate that hormones or factors related to oral contraceptive use are acting early in RA-related immune dysregulation to reduce risk for formation of autoantibodies.

Infection and mucosal inflammation and the initiation of RA-related autoimmunity

RA-related autoimmunity has been associated with multiple organisms as well as tissue inflammation and injury at sites other than the joints, including the lungs, genitourinary tract and oral cavity/periodontal region. It has been typically thought that such infections and tissue inflammation are secondarily due to immunosuppression or autoimmune-related injury. However, the growing understanding of the preclinical phases of RA development71, and recent findings show that circulating RF and ACPA elevations are present in the absence of synovitis on knee synovial biopsy provide support for the concept that RA may be generated outside of the joints.111 Therefore, it may be that the infections that are associated with autoantibody formation, and inflammation at these extra-articular sites, are providing clues as to etiologic factors and sites that are driving the generation of initial RA-related autoimmunity.

As a specific example of infection and mucosal inflammation in association with RA, recent data has shown an association between established RA and periodontal disease.112115 The causal direction remains uncertain but recent discoveries suggest that gum inflammation and in particular infection with organisms such as Porphyromonas gingivalis (P ging) may be important in the pathophysiology of RA development.113115 The current hypothesis is that P. ging-mediated citrullination of human peptides may be responsible for the initial breakdown in self-tolerance that leads to the development of RA-related autoimmunity.112,116 This is an intriguing concept, but it, as well as the role of multiple other infectious organisms and inflammation at other mucosal sites (including the respiratory tract, gut and genito-urinary tract) that have been implicated in the pathogenesis of RA, needs further exploration.117125 Importantly, such studies investigating the roles of infectious and mucosal factors in RA need to be performed in prospective study of the preclinical phases of RA development in order to identify the true temporal as well as mechanistic relationships between infections and mucosal inflammation in the early pathogenesis of RA.

Dietary factors

Dietary intake of vitamin D, antioxidants, fish and protein and iron may be inversely associated with RA risk although the data is mixed.73,126137 Vitamin D has been implicated as an etiologic and therapeutic factor in several autoimmune diseases including multiple sclerosis,138,139 Type 1 diabetes140, and SLE.141 Vitamin D has pleotropic effects on the immune system, inhibiting pro-inflammatory cytokines, upregulating anti-inflammatory cytokines,142 and regulating the innate and adaptive immune system through the vitamin D receptor.140 Vitamin D prevents the development of inflammatory arthritis in collagen induced mouse models.143 One study found a strong protective effect of high vitamin D intake in diminishing RA risk showing inverse associations for dietary and supplemental vitamin D73; another study revealed no association136. Furthermore, in a study of ~1200 healthy individuals at risk for future RA because of genetic factors (HLA DR4 allele enrichment and/or family history of RA), 76 of whom were positive for the RA-related autoantibodies anti-CCP and/or RF, there was no association of autoantibody positivity with plasma vitamin D levels.144 Studies show an inverse relationship between higher vitamin D and lower disease activity in RA after diagnosis.145,146

Marine omega-3 fatty acids exert effects through the leukotriene and prostaglandin pathways, decreasing inflammatory mediators, and have multiple known anti-inflammatory properties.147 Case-control and cohort studies reported a modest protective effect of fish intake on RA risk.127,128,133,148 Consumption of fish has also been shown to improve RA symptoms and delay RA progression.149,150

Antioxidants

Observational case-control and cohort studies suggest that antioxidants may protect against the development of RA but the results for individual antioxidants are conflicting.128132,137,151153 The Iowa Women’s Health Study demonstrated an inverse association for β-cryptoxanthin and supplemental zinc, but only a weak inverse association for vitamin C, and no association for vitamin E and risk of RA.129 In NOAR, Vitamin C was inversely associated with the risk of IA, but β-cryptoxanthin and zeaxanthin antioxidants showed only weak inverse associations after adjustment for Vitamin C.130,131 Data from the NHS did not support any prospective association for antoxidants including vitamin C, vitamin E and β-cryptoxanthin.137 Furthermore, data from a case-control study128 and a randomized controlled trial135 did not support an effect of Vitamin E on RA risk.

Protein and iron

In NOAR higher intakes of red meat and protein were associated with risk of IA while iron, another nutrient component of meat, showed no association.130,132 However, no clear associations were observed between dietary protein, iron or meats, including red meat, and risk of RA in the NHS cohort.134

Alcohol

Case-control studies have suggested that alcohol consumption may decrease the risk for RA and RA progression.49,50,154 Any alcohol vs. no alcohol use was associated with lower odds of seropositive RA in several case-control studies.17,50,155 A dose dependent protected effect was demonstrated in two case control studies in which alcohol consumption was based on patient questionnaires regarding alcohol consumption in the previous week, previous habitual consumption prior to RA onset or consumption 10 years before inclusion.49 Furthermore, there was evidence for alcohol*HLA-SE interaction. However, there was no association between alcohol and RA in the IWHS, a prospective cohort study.156 In the NHS, moderate alcohol intake (3–6 drinks per week) was associated with reduction in plasma biomarkers of inflammation, including CRP, sTNFR2, and IL-6, among women with pre-RA where blood was collected up to 12 years prior to first symptoms of RA suggesting an effect of alcohol on inflammation during the asymptomatic phases.157

There are several possible mechanisms for the inverse association of alcohol consumption with RA. Moderate alcohol consumption may be associated with reduced levels of inflammatory biomarkers reflective of underlying improved alterations of inflammatory pathways.158 Alcohol has also been shown to diminish the response to antigens in animals as well as in humans,159,160 and to suppress the synthesis of proinflammatory cytokines and chemokines, such as TNFα, IL-6 and IL-8 both in vivo and in vitro in alveolar macrophages and human blood monocytes.161,162

Inhaled Particulate Air Pollution

While the first cases of RA may date back thousands of years, the prevalence of RA appears to have increased in Europe in the 1800s.163,164 This time period is associated with the introduction of tobacco from the New World and increased popularity of smoking and it also coincides with the Industrial Revolution and the advent of air pollution in Europe.165 In the EIRA cohort, lower socioeconomic status is associated with an increased risk of RA.166 This association remains strong after adjustment for cigarette smoking, suggesting the existence of an important environmental or lifestyle factor associated with lower socioeconomic status. This finding is particularly interesting given that exposure to air pollution is often much higher for those in lower socioeconomic classes.167,168 Our evolving understanding of RA pathogenesis suggests that inhaled particulate matter may induce local lung inflammation as well as through systemic inflammation. Indirect support of this hypothesis comes from the observation that air pollution has been clearly linked with other diseases of local lung and systemic inflammation, including asthma169 and chronic bronchitis170,171, cardiovascular disease172174, lung172,175,176 and laryngeal177 cancers and increased all-cause mortality.178180 Studies from the NHS reported a higher relative risk of RA in the Northeast and upper Mid-West regions of the US, regions with high air pollution levels.181,182 Further, data suggests a higher RA risk in women who live closest to major road, a proxy for traffic pollution.47

Family history

Twin studies demonstrate a high concordance of RA between monozygotic compared with dizygotic twins.183 In a Swedish study involving 47,361 subjects with RA, standardized incidence ratios (SIR) were calculated as relative risk (RR) of RA in family members of RA patients as compared with RR in those with no affected family members.53 Standardized Incidence Ratios (SIRs) for RA were 3.0 in offspring of RA-affected parents, 4.6 in siblings, 9.3 in multiplex families (both parent and sibling), and 6.5 in twins. The 3- to 9- fold increased familial risk of RA suggests strong influence of genetic or shared environmental effects or both.

Interactions between environmental and genetic factors and RA risk

There are several potential mechanisms by-which gene-environment interactions may ‘trigger’ RA-related autoimmunity. A gene–environment interaction between HLA-SE and smoking was first described in landmark studies led by Klareskog and colleagues.13,77 This work has demonstrated that interaction between these two factors was strongly associated with specific phenotypes of ACPA+ and RF+, whereas no such association was seen for the risk of developing seronegative RA. Furthermore, the additive nature of the interaction suggested a biologic pathway to disease onset. Subsequent studies replicated and expanded upon these findings using two different methods: case–control analyses comparing RA cases stratified according to autoantibody phenotype with healthy controls5 and case-only analyses comparing autoantibody-positive RA cases to autoantibody-negative RA cases, without involving healthy controls.184,185 Gene environment interaction between HLA-SE in a case-control study was strongest for autoantibody positive RA with significant multiplicative interaction.58 Further, there was a dose response in the gene-environment interactions for both allele dose and smoking does with highest risk among heavy smokers with double copy HLA-SE. Gene-environment interactions between GSTT1 or GSM1 null polymorphisms (resulting in absence of the GSTT1 or GSTM1 enzymes that detoxify cigarette smoke), and the drug-metabolizing enzyme N-acetyletransferase-2 (NAT2) and smoking further supports the synergistic effect smoking and genetics on risk for RA.186189

In an attempt to identify specific mechanistic pathways by-which exposure to tobacco smoke may induce RA-related autoimmunity, investigators have also examined broncho-alveolar lavage fluid from healthy non-smokers, healthy smokers and smokers with concomitant inflammatory lung diseases (such as sarcoidosis and Langerhan’s cell histiocytosis).13,79 They found that smoking increases PAD2 enzyme expression in human lungs and was associated with increased proportion of citrulline-positive BAL cells (mainly alveolar macrophages), whereas citrullinated cells were not found in nonsmokers.

Overall, this research has led to the hypothesis that smoking induces citrullination, with subsequent immune reactions to citrullinated antigens occurring in HLA-SE-positive individuals. This hypothesis is strengthened by the demonstration in HLA-DRB1* 0401 transgenic mice that citrullination of certain peptides renders them more prone to bind to HLA class II molecules with the HLA-SE, and to trigger a strong immune response to citrullinated self-antigens.190 If similar mechanisms are occurring humans to drive the development of RA, based on the ‘phased’ model of RA development (Figure 1), the gene-environmental interactions that may lead to RA-related autoimmunity are likely occurring some time prior to the onset of symptomatic joint disease. However, the mechanisms as well as the specific sequence of gene-environment interactions such as smoking, the generation of citrullinated proteins and subsequent autoimmunity to those proteins, that lead to symptomatic RA are unknown. For example, it may be that some factor other than smoking initially interacts with HLA alleles to generate autoantibodies to citrullinated proteins, and that smoking is a permissive factor that later drives the development of symptomatic RA rather than the initiation of autoimmunity. To fully understand these issues, the development of RA needs to be explored in prospective studies that can in real-time evaluate the relationships between gene-environment interactions and the development of RA-related autoimmunity.

Pathologic roles for autoantibodies in RA

Despite their strong association with RA, it is unclear what precise role(s) for autoantibodies including RF and ACPAs have in the pathogenesis of RA, although it will be important to define these roles especially given the data discussed above associating the development of seropositive RA with gene-environmental interactions.5,13,5557,191 However, numerous studies suggest that autoantibodies are likely a key aspect of the development of RA.192,193 In particular, the association of RF and ACPAs with more severe RA (or arthritis in non-RA conditions such as SLE) suggests that these autoantibodies are important contributors to joint disease.63,192195 Also, in animal models of arthritis, infusion of antibodies to citrullinated proteins leads to more severe arthritis suggesting these autoantibodies play a direct pathogenic role in disease development.196 In addition, recent work demonstrating in both animal models as well as humans that circulating immune complexes containing ACPAs and citrullinated fibrinogen activate inflammation through interactions with Toll-like receptor 4 provides a potential mechanism by which circulating ACPAs may induce joint-specific inflammation – a process that is enhanced if RF is also present.197,198 Finally, as discussed above, the findings in multiple human studies of elevated RF and ACPAs prior to the onset of RA suggest that autoimmunity to citrullinated proteins is an important factor in the development of RA in the period prior to the appearance of symptomatic synovitis. In sum, these data suggest that autoantibodies are an important aspect of the pathogenesis of RA; therefore, the area of gene-environment interactions that may lead to the development of these autoantibodies is a fertile ground for investigations in the pathogenesis of RA.

Animal studies linking environmental factors to development of autoantibodies

Emerging data from animal models of disease that have explored the role of environmental factors such as smoking in the pathogenesis of IA may provide insight into specific mechanism by-which environmental exposures may ‘trigger’ RA-related autoimmunity. In particular, in a murine model of collagen-induced arthritis, Okamoto and colleagues induced IA by exposing the mice nasally to cigarette smoke condensate.199 However, other studies have shown that exposure to cigarette smoke delayed collagen-induced arthritis in mice, perhaps due to immunosuppressive constituents of cigarette smoke.200 Therefore, additional work is needed to develop models that can help explain the pathophysiologic mechanisms by-which exposure to environmental risk factors such as cigarette smoke may trigger RA. As for potential infectious triggers of RA, multiple animal models of collagen-induced arthritis have shown more severe arthritis in the setting of environments where organisms are present.201,202 Furthermore, in 2011, Kinloch and colleagues reported in a HLA DR4-transgenic murine model of arthritis that immunization with both citrullinated and uncitrullinated forms of enolase from P ging led to autoantibodies to the citrullinated mammalian form of this enzyme as well as arthritis.203 This latter discovery needs further work to determine the direct applicability to human disease; however, it does support a hypothesis that bacterial factors may be an important part of development of RA.

Remaining questions and future directions

The identification of autoantibodies and cytokines present many years prior to RA onset, and the association of many environmental factors with RA development provides an exciting opportunity to intervene during the pre-clinical phase to prevent the development of symptomatic RA. However, it is critical to understand the critical exposure windows to design interventions, especially if such interventions are to include modification of environmental exposures. Further research into the environment and gene-environment determinants of RA risk will provide important data for individualized studies in order to target potentially toxic therapy at individuals of highest risk, with such data preferable obtained through detailed, prospective studies of the natural history of RA so that we can understand the precise temporal as well as mechanistic relationships between genetic and environmental factors and the development of RA-related autoimmunity. The concept that RA is initiated at some mucosal site distal to the joints, potentially in relationship to environmental factors such as smoking, alcohol or infection, is fascinating and may lead to a significant break-through in our understanding of how this disease develops, and needs additional study.

The ability to accurately predict an individual’s risk of developing clinical RA would be an enormous advance in this area, enabling risk factor modification and earlier introduction of effective therapies to abrogate the destruction and disability of this disease. Thus predictive modeling incorporating RA genetic susceptibility alleles, hormonal, environmental, and behavioral risk factors, and their interactions is a key step in the progress towards an RA prevention clinical trial.

KEY POINTS.

  • Individuals with inherited genetic risk factors who are exposed to environmental triggers are at higher risk of RA.

  • Multiple environmental factors including exposure to tobacco smoke, occupational exposures, hormones, infections and dietary factors are associated with RA risk.

  • Circulating RA-related autoimmunity is evident in some patients prior to the appearance of the first joint symptoms and clinical findings of RA. This asymptomatic phase of disease development suggests that important gene and environmental interactions leading to initiation of RA-related autoimmunity occur long prior to the onset of clinically-apparent disease.

  • Natural history studies of the asymptomatic phase of RA development are needed to determine the mechanistic role(s) that environmental factors play in the initiation of RA.

  • Animal studies link autoantibodies to RA pathogenesis and going forward may provide insight to the mechanisms of environmental and genetic factors in the pathogenesis of RA.

Acknowledgments

Dr. Karlson’s work was supported by grants Supported by NIH grants AR049880, AR052403, and AR047782. Dr. Deane’s work was supported by NIH grants AR051394, AR07534, AR051461, AI50864, the American College of Rheumatology Research and Education Foundation’s Within Our Reach Program, and the Walter S. and Lucienne Driskill Foundation.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Disclosures: The authors have nothing to disclose.

References

  • 1.Majka DS, Deane KD, Parrish LA, et al. Duration of preclinical rheumatoid arthritis-related autoantibody positivity increases in subjects with older age at time of disease diagnosis. Annals of the rheumatic diseases. 2008;67:801–807. doi: 10.1136/ard.2007.076679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Deane KD, O'Donnell CI, Hueber W, et al. The number of elevated cytokines and chemokines in preclinical seropositive rheumatoid arthritis predicts time to diagnosis in an age-dependent manner. Arthritis and rheumatism. 2010;62:3161–3172. doi: 10.1002/art.27638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Rantapaa-Dahlqvist S, de Jong BA, Berglin E, et al. Antibodies against cyclic citrullinated peptide and IgA rheumatoid factor predict the development of rheumatoid arthritis. Arthritis and rheumatism. 2003;48:2741–2749. doi: 10.1002/art.11223. [DOI] [PubMed] [Google Scholar]
  • 4.Nielen MM, van Schaardenburg D, Reesink HW, et al. Specific autoantibodies precede the symptoms of rheumatoid arthritis: a study of serial measurements in blood donors. Arthritis and rheumatism. 2004;50:380–386. doi: 10.1002/art.20018. [DOI] [PubMed] [Google Scholar]
  • 5.Chibnik LB, Mandl LA, Costenbader KH, Schur PH, Karlson EW. Comparison of threshold cutpoints and continuous measures of anti-cyclic citrullinated peptide antibodies in predicting future rheumatoid arthritis. J Rheumatol. 2009;36:706–711. doi: 10.3899/jrheum.080895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kokkonen H, Mullazehi M, Berglin E, et al. Antibodies of, IgG, IgA and IgM isotypes against cyclic citrullinated peptide precede the development of rheumatoid arthritis. Arthritis research &therapy. 2010;13:R13. doi: 10.1186/ar3237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kokkonen H, Soderstrom I, Rocklov J, Hallmans G, Lejon K, Rantapaa Dahlqvist S. Up-regulation of cytokines and chemokines predates the onset of rheumatoid arthritis. Arthritis and rheumatism. 2010;62:383–391. doi: 10.1002/art.27186. [DOI] [PubMed] [Google Scholar]
  • 8.Jorgensen KT, Wiik A, Pedersen M, et al. Cytokines, autoantibodies and viral antibodies in premorbid and postdiagnostic sera from patients with rheumatoid arthritis: case-control study nested in a cohort of Norwegian blood donors. Annals of the rheumatic diseases. 2008;67:860–866. doi: 10.1136/ard.2007.073825. [DOI] [PubMed] [Google Scholar]
  • 9.Arnett FC, Edworthy SM, Bloch DA, et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis and rheumatism. 1988;31:315–324. doi: 10.1002/art.1780310302. [DOI] [PubMed] [Google Scholar]
  • 10.Aletaha D, Neogi T, Silman AJ, et al. Rheumatoid arthritis classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Arthritis and rheumatism. 2010;62:2569–2581. doi: 10.1002/art.27584. [DOI] [PubMed] [Google Scholar]
  • 11.Leslie D, Lipsky P, Notkins AL. Autoantibodies as predictors of disease. J Clin Invest. 2001;108:1417–1422. doi: 10.1172/JCI14452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Arbuckle MR, McClain MT, Rubertone MV, et al. Development of autoantibodies before the clinical onset of systemic lupus erythematosus. N Engl J Med. 2003;349:1526–1533. doi: 10.1056/NEJMoa021933. [DOI] [PubMed] [Google Scholar]
  • 13.Klareskog L, Stolt P, Lundberg K, et al. A new model for an etiology of rheumatoid arthritis: Smoking may trigger HLA-DR (shared epitope)-restricted immune reactions to autoantigens modified by citrullination. Arthritis and rheumatism. 2006;54:38–46. doi: 10.1002/art.21575. [DOI] [PubMed] [Google Scholar]
  • 14.Vessey MP, Villard-Mackintosh L, Yeates D. Oral contraceptives, cigarette smoking and other factors in relation to arthritis. Contraception. 1987;35:457–464. doi: 10.1016/0010-7824(87)90082-5. [DOI] [PubMed] [Google Scholar]
  • 15.Hernandez Avila M, Liang MH, Willett WC, et al. Reproductive factors, smoking, and the risk for rheumatoid arthritis. Epidemiology. 1990;1:285–291. doi: 10.1097/00001648-199007000-00005. [DOI] [PubMed] [Google Scholar]
  • 16.Hernandez-Avila M, Liang MH, Willett WC, et al. Exogenous sex hormones and the risk of rheumatoid arthritis. Arthritis and rheumatism. 1990;33:947–953. doi: 10.1002/art.1780330705. [DOI] [PubMed] [Google Scholar]
  • 17.Hazes JM, Dijkmans BA, Vandenbroucke JP, de Vries RR, Cats A. Lifestyle and the risk of rheumatoid arthritis: cigarette smoking and alcohol consumption. AnnRheumDis. 1990;49:980–982. doi: 10.1136/ard.49.12.980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Heliovaara M, Aho K, Aromaa A, Knekt P, Reunanen A. Smoking and risk of rheumatoid arthritis. J Rheumatol. 1993;20:1830–1835. [see comments] [PubMed] [Google Scholar]
  • 19.Voigt LF, Koepsell TD, Nelson JL, Dugowson CE, Daling JR. Smoking, obesity, alcohol consumption, and the risk of rheumatoid arthritis. Epidemiology. 1994;5:525–532. [PubMed] [Google Scholar]
  • 20.Symmons DP, Bankhead CR, Harrison BJ, et al. Blood transfusion, smoking, and obesity as risk factors for the development of rheumatoid arthritis: Results from a primary care-based incident case-control study in Norfolk, England. Arthritis and rheumatism. 1997;40:1955–1961. doi: 10.1002/art.1780401106. [DOI] [PubMed] [Google Scholar]
  • 21.Karlson EW, Lee IM, Cook NR, Manson JE, Buring JE, Hennekens CH. A retrospective cohort study of cigarette smoking and risk of rheumatoid arthritis in female health professionals. Arthritis and rheumatism. 1999;42:910–917. doi: 10.1002/1529-0131(199905)42:5<910::AID-ANR9>3.0.CO;2-D. [DOI] [PubMed] [Google Scholar]
  • 22.Uhlig T, Hagen KB, Kvien TK. Current tobacco smoking, formal education, and the risk of rheumatoid arthritis. J Rheumatol. 1999;26:47–54. [PubMed] [Google Scholar]
  • 23.Kallberg H, Ding B, Padyukov L, et al. Smoking is a major preventable risk factor for rheumatoid arthritis: estimations of risks after various exposures to cigarette smoke. Annals of the rheumatic diseases. 2011;70:508–511. doi: 10.1136/ard.2009.120899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Mikuls TR, Saag KG, Criswell LA, et al. Mortality risk associated with rheumatoid arthritis in a prospective cohort of older women: results from the Iowa Women's Health Study. Ann Rheum Dis. 2002;61:994–999. doi: 10.1136/ard.61.11.994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Krishnan E, Sokka T, Hannonen P. Smoking-gender interaction and risk for rheumatoid arthritis. Arthritis research & therapy. 2003;5:R158–R162. doi: 10.1186/ar750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Stolt P, Bengtsson C, Nordmark B, et al. Quantification of the influence of cigarette smoking on rheumatoid arthritis: results from a population based case-control study, using incident cases. Annals of the rheumatic diseases. 2003;62:835–841. doi: 10.1136/ard.62.9.835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Costenbader KH, Feskanich D, Mandl LA, Karlson EW. Smoking intensity, duration, and cessation, and the risk of rheumatoid arthritis in women. Am J Med. 2006;119:503–511. doi: 10.1016/j.amjmed.2005.09.053. [DOI] [PubMed] [Google Scholar]
  • 28.Doran MF, Crowson CS, O'Fallon WM, Gabriel SE. The effect of oral contraceptives and estrogen replacement therapy on the risk of rheumatoid arthritis: a population based study. J Rheumatol. 2004;31:207–213. [PubMed] [Google Scholar]
  • 29.Spector TD, Roman E, Silman AJ. The pill, parity, and rheumatoid arthritis. Arthritis Rheum. 1990;33:782–789. doi: 10.1002/art.1780330604. [DOI] [PubMed] [Google Scholar]
  • 30.Brennan P, Bankhead C, Silman A, Symmons D. Oral contraceptives and rheumatoid arthritis: results from a primary care-based incident case-control study. SeminArthritis Rheum. 1997;26:817–823. doi: 10.1016/s0049-0172(97)80025-x. [DOI] [PubMed] [Google Scholar]
  • 31.Hazes JM, Dijkmans BC, Vandenbroucke JP, de Vries RR, Cats A. Reduction of the risk of rheumatoid arthritis among women who take oral contraceptives. Arthritis Rheum. 1990;33:173–179. doi: 10.1002/art.1780330204. [see comments] [DOI] [PubMed] [Google Scholar]
  • 32.Jorgensen C, Picot MC, Bologna C, Sany J. Oral contraception, parity, breast feeding, and severity of rheumatoid arthritis. Annals of the rheumatic diseases. 1996;55:94–98. doi: 10.1136/ard.55.2.94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Vandenbroucke JP, Valkenburg HA, Boersma JW, et al. Oral contraceptives and rheumatoid arthritis: further evidence for a preventive effect. Lancet. 1982;2:839–842. doi: 10.1016/s0140-6736(82)90809-1. [DOI] [PubMed] [Google Scholar]
  • 34.Vandenbroucke JP, Witteman JC, Valkenburg HA, et al. Noncontraceptive hormones and rheumatoid arthritis in perimenopausal and postmenopausal women. Jama. 1986;255:1299–1303. [PubMed] [Google Scholar]
  • 35.Wingrave SJ, Kay CR. Reduction in incidence of rheumatoid arthritis associated with oral contraceptives. Lancet. 1978;1:569–571. [PubMed] [Google Scholar]
  • 36.Walitt B, Pettinger M, Weinstein A, et al. Effects of postmenopausal hormone therapy on rheumatoid arthritis: the women's health initiative randomized controlled trials. Arthritis and rheumatism. 2008;59:302–310. doi: 10.1002/art.23325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Merlino LA, Cerhan JR, Criswell LA, Mikuls TR, Saag KG. Estrogen and other female reproductive risk factors are not strongly associated with the development of rheumatoid arthritis in elderly women. Semin Arthritis Rheum. 2003;33:72–82. doi: 10.1016/s0049-0172(03)00084-2. [DOI] [PubMed] [Google Scholar]
  • 38.Karlson EW, Mandl LA, Hankinson SE, Grodstein F. Do breast-feeding and other reproductive factors influence future risk of rheumatoid arthritis? Results from the Nurses' Health Study. Arthritis and rheumatism. 2004;50:3458–3467. doi: 10.1002/art.20621. [DOI] [PubMed] [Google Scholar]
  • 39.Pikwer M, Bergstrom U, Nilsson JA, Jacobsson L, Berglund G, Turesson C. Breast feeding, but not use of oral contraceptives, is associated with a reduced risk of rheumatoid arthritis. Annals of the rheumatic diseases. 2009;68:526–530. doi: 10.1136/ard.2007.084707. [DOI] [PubMed] [Google Scholar]
  • 40.Berglin E, Kokkonen H, Einarsdottir E, Agren A, Rantapaa Dahlqvist S. Influence of female hormonal factors, in relation to autoantibodies and genetic markers, on the development of rheumatoid arthritis in northern Sweden: a case-control study. Scandinavian journal of rheumatology. 2010;39:454–460. doi: 10.3109/03009741003742763. [DOI] [PubMed] [Google Scholar]
  • 41.Pikwer M, Bergstrom U, Nilsson JA, Jacobsson L, Turesson C. Early menopause is an independent predictor of rheumatoid arthritis. Annals of the rheumatic diseases. 2012;71:378–381. doi: 10.1136/ard.2011.200059. [DOI] [PubMed] [Google Scholar]
  • 42.Stolt P, Kallberg H, Lundberg I, Sjogren B, Klareskog L, Alfredsson L. Silica exposure is associated with increased risk of developing rheumatoid arthritis: results from the Swedish EIRA study. Annals of the rheumatic diseases. 2005;64:582–586. doi: 10.1136/ard.2004.022053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Sluis-Cremer GK, Hessel PA, Hnizdo E, Churchill AR. Relationship between silicosis and rheumatoid arthritis. Thorax. 1986;41:596–601. doi: 10.1136/thx.41.8.596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Turner S, Cherry N. Rheumatoid arthritis in workers exposed to silica in the pottery industry. Occup Environ Med. 2000;57:443–447. doi: 10.1136/oem.57.7.443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Klockars M, Koskela RS, Jarvinen E, Kolari PJ, Rossi A. Silica exposure and rheumatoid arthritis: a follow up study of granite workers 1940-81. Br Med J (Clin Res Ed) 1987;294:997–1000. doi: 10.1136/bmj.294.6578.997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Steenland K, Sanderson W, Calvert GM. Kidney disease and arthritis in a cohort study of workers exposed to silica. Epidemiology. 2001;12:405–412. doi: 10.1097/00001648-200107000-00010. [DOI] [PubMed] [Google Scholar]
  • 47.Hart JE, Laden F, Puett RC, Costenbader KH, Karlson EW. Exposure to traffic pollution and increased risk of rheumatoid arthritis. Environmental health perspectives. 2009;117:1065–1069. doi: 10.1289/ehp.0800503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.de Pablo P, Chapple IL, Buckley CD, Dietrich T. Periodontitis in systemic rheumatic diseases. Nature reviews. 2009;5:218–224. doi: 10.1038/nrrheum.2009.28. [DOI] [PubMed] [Google Scholar]
  • 49.Kallberg H, Jacobsen S, Bengtsson C, et al. Alcohol consumption is associated with decreased risk of rheumatoid arthritis: results from two Scandinavian case-control studies. Annals of the rheumatic diseases. 2009;68:222–227. doi: 10.1136/ard.2007.086314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Pedersen M, Jacobsen S, Klarlund M, et al. Environmental risk factors differ between rheumatoid arthritis with and without auto-antibodies against cyclic citrullinated peptides. Arthritis research & therapy. 2006;8:R133. doi: 10.1186/ar2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Jacobsson LT, Jacobsson ME, Askling J, Knowler WC. Perinatal characteristics and risk of rheumatoid arthritis. BMJ (Clinical research ed. 2003;326:1068–1069. doi: 10.1136/bmj.326.7398.1068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Mandl LA, Costenbader KH, Simard JF, Karlson EW. Is birthweight associated with risk of rheumatoid arthritis? Data from a large cohort study. Annals of the rheumatic diseases. 2009;68:514–518. doi: 10.1136/ard.2007.080937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Hemminki K, Li X, Sundquist J, Sundquist K. Familial associations of rheumatoid arthritis with autoimmune diseases and related conditions. Arthritis and rheumatism. 2009;60:661–668. doi: 10.1002/art.24328. [DOI] [PubMed] [Google Scholar]
  • 54.Stahl EA, Raychaudhuri S, Remmers EF, et al. Genome-wide association study meta-analysis identifies seven new rheumatoid arthritis risk loci. Nature genetics. 2010;42:508–514. doi: 10.1038/ng.582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Costenbader KH, Chang SC, De Vivo I, Plenge R, Karlson EW. Genetic polymorphisms in PTPN22, PADI-4, and CTLA-4 and risk for rheumatoid arthritis in two longitudinal cohort studies: evidence of gene-environment interactions with heavy cigarette smoking. Arthritis research & therapy. 2008;10:R52. doi: 10.1186/ar2421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Karlson EW, Chibnik LB, Kraft P, et al. Cumulative association of 22 genetic variants with seropositive rheumatoid arthritis risk. Annals of the rheumatic diseases. 2010;69:1077–1085. doi: 10.1136/ard.2009.120170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Mahdi H, Fisher BA, Kallberg H, et al. Specific interaction between genotype, smoking and autoimmunity to citrullinated alpha-enolase in the etiology of rheumatoid arthritis. Nature genetics. 2009;41:1319–1324. doi: 10.1038/ng.480. [DOI] [PubMed] [Google Scholar]
  • 58.Karlson EW, Chang SC, Cui J, et al. Gene-environment interaction between HLA-DRB1 shared epitope and heavy cigarette smoking in predicting incident rheumatoid arthritis. Annals of the rheumatic diseases. 2010;69:54–60. doi: 10.1136/ard.2008.102962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.del Puente A, Knowler WC, Pettitt DJ, Bennett PH. The incidence of rheumatoid arthritis is predicted by rheumatoid factor titer in a longitudinal population study. Arthritis and rheumatism. 1988;31:1239–1244. doi: 10.1002/art.1780311004. [DOI] [PubMed] [Google Scholar]
  • 60.Del Puente A, Knowler WC, Pettitt DJ, Bennett PH. High incidence and prevalence of rheumatoid arthritis in Pima Indians. Am J Epidemiol. 1989;129:1170–1178. doi: 10.1093/oxfordjournals.aje.a115238. [DOI] [PubMed] [Google Scholar]
  • 61.Silman AJ, Hennessy E, Ollier B. Incidence of rheumatoid arthritis in a genetically predisposed population. Br J Rheumatol. 1992;31:365–368. doi: 10.1093/rheumatology/31.6.365. [DOI] [PubMed] [Google Scholar]
  • 62.Aho K, Heliovaara M, Maatela J, Tuomi T, Palosuo T. Rheumatoid factors antedating clinical rheumatoid arthritis. J Rheumatol. 1991;18:1282–1284. [PubMed] [Google Scholar]
  • 63.Aho K, Palusuo T, Kurki P. Marker antibodies of rheumatoid arthritis: diagnostic and pathogenetic implications. Semin Arthritis Rheum. 1994;23:379–387. doi: 10.1016/0049-0172(94)90088-4. [DOI] [PubMed] [Google Scholar]
  • 64.Aho K, Palosuo T, Heliovaara M, Knekt P, Alha P, von Essen R. Antifilaggrin antibodies within "normal" range predict rheumatoid arthritis in a linear fashion. J Rheumatol. 2000;27:2743–2746. [PubMed] [Google Scholar]
  • 65.Berglin E, Padyukov L, Sundin U, et al. A combination of autoantibodies to cyclic citrullinated peptide (CCP) and HLA-DRB1 locus antigens is strongly associated with future onset of rheumatoid arthritis. Arthritis research & therapy. 2004;6:R303–R308. doi: 10.1186/ar1187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Nielen MM, van Schaardenburg D, Reesink HW, et al. Simultaneous development of acute phase response and autoantibodies in preclinical rheumatoid arthritis. Annals of the rheumatic diseases. 2006;65:535–537. doi: 10.1136/ard.2005.040659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Rantapaa-Dahlqvist S, Boman K, Tarkowski A, Hallmans G. Up regulation of monocyte chemoattractant protein-1 expression in anti-citrulline antibody and immunoglobulin M rheumatoid factor positive subjects precedes onset of inflammatory response and development of overt rheumatoid arthritis. Annals of the rheumatic diseases. 2007;66:121–123. doi: 10.1136/ard.2006.057331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Bos WH, Ursum J, de Vries N, et al. The role of the shared epitope in arthralgia with anti-cyclic citrullinated peptide antibodies (anti-CCP), and its effect on anti-CCP levels. Annals of the rheumatic diseases. 2008;67:1347–1350. doi: 10.1136/ard.2008.089953. [DOI] [PubMed] [Google Scholar]
  • 69.Bos WH, Wolbink GJ, Boers M, et al. Arthritis development in patients with arthralgia is strongly associated with anti-citrullinated protein antibody status: a prospective cohort study. Annals of the rheumatic diseases. 2010;69:490–494. doi: 10.1136/ard.2008.105759. [DOI] [PubMed] [Google Scholar]
  • 70.Majka DS, Holers VM. Can we accurately predict the development of rheumatoid arthritis in the preclinical phase? Arthritis and rheumatism. 2003;48:2701–2705. doi: 10.1002/art.11224. [DOI] [PubMed] [Google Scholar]
  • 71.Deane KD, Norris JM, Holers VM. Preclinical rheumatoid arthritis: identification, evaluation, and future directions for investigation. Rheum Dis Clin North Am. 2010;36:213–241. doi: 10.1016/j.rdc.2010.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Chouinard E, Walter S. Recall bias in case-control studies: an empirical analysis and theoretical framework. Journal of clinical epidemiology. 1995;48:245–254. doi: 10.1016/0895-4356(94)00132-a. [DOI] [PubMed] [Google Scholar]
  • 73.Merlino LA, Curtis J, Mikuls TR, Cerhan JR, Criswell LA, Saag KG. Vitamin D intake is inversely associated with rheumatoid arthritis: results from the Iowa Women's Health Study. Arthritis and rheumatism. 2004;50:72–77. doi: 10.1002/art.11434. [DOI] [PubMed] [Google Scholar]
  • 74.Symmons DP, Silman AJ. The Norfolk Arthritis Register (NOAR) Clinical and experimental rheumatology. 2003;21:S94–S99. [PubMed] [Google Scholar]
  • 75.Kolfenbach JR, Deane KD, Derber LA, et al. A prospective approach to investigating the natural history of preclinical rheumatoid arthritis (RA) using first-degree relatives of probands with RA. Arthritis and rheumatism. 2009;61:1735–1742. doi: 10.1002/art.24833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.El-Gabalawy HS, Robinson DB, Hart D, et al. Immunogenetic risks of anti-cyclical citrullinated peptide antibodies in a North American Native population with rheumatoid arthritis and their first-degree relatives. J Rheumatol. 2009;36:1130–1135. doi: 10.3899/jrheum.080855. [DOI] [PubMed] [Google Scholar]
  • 77.Padyukov L, Silva C, Stolt P, Alfredsson L, Klareskog L. A gene-environment interaction between smoking and shared epitope genes in HLA-DR provides a high risk of seropositive rheumatoid arthritis. Arthritis and rheumatism. 2004;50:3085–3092. doi: 10.1002/art.20553. [DOI] [PubMed] [Google Scholar]
  • 78.Stolt P, Yahya A, Bengtsson C, et al. Silica exposure among male current smokers is associated with a high risk of developing ACPA-positive rheumatoid arthritis. Annals of the rheumatic diseases. 2010;69:1072–1076. doi: 10.1136/ard.2009.114694. [DOI] [PubMed] [Google Scholar]
  • 79.Makrygiannakis D, Hermansson M, Ulfgren AK, et al. Smoking increases peptidylarginine deiminase 2 enzyme expression in human lungs and increases citrullination in BAL cells. Annals of the rheumatic diseases. 2008;67:1488–1492. doi: 10.1136/ard.2007.075192. [DOI] [PubMed] [Google Scholar]
  • 80.Verpoort KN, Cheung K, Ioan-Facsinay A, et al. Fine specificity of the anti-citrullinated protein antibody response is influenced by the shared epitope alleles. Arthritis and rheumatism. 2007;56:3949–3952. doi: 10.1002/art.23127. [DOI] [PubMed] [Google Scholar]
  • 81.Snir O, Widhe M, von Spee C, et al. Multiple antibody reactivities to citrullinated antigens in sera from patients with rheumatoid arthritis: association with HLA-DRB1 alleles. Annals of the rheumatic diseases. 2009;68:736–743. doi: 10.1136/ard.2008.091355. [DOI] [PubMed] [Google Scholar]
  • 82.Feitsma AL, van der Voort EI, Franken KL, et al. Identification of citrullinated vimentin peptides as T cell epitopes in HLA-DR4-positive patients with rheumatoid arthritis. Arthritis and rheumatism. 2010;62:117–125. doi: 10.1002/art.25059. [DOI] [PubMed] [Google Scholar]
  • 83.Bhatia SS, Majka DS, Kittelson JM, et al. Rheumatoid factor seropositivity is inversely associated with oral contraceptive use in women without rheumatoid arthritis. Annals of the rheumatic diseases. 2007;66:267–269. doi: 10.1136/ard.2006.060004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Hughes DA, Haslam PL, Townsend PJ, Turner-Warwick M. Numerical and functional alterations in circulatory lymphocytes in cigarette smokers. Clin Exp Immunol. 1985;61:459–466. [PMC free article] [PubMed] [Google Scholar]
  • 85.Robbins CS, Dawe DE, Goncharova SI, et al. Cigarette smoke decreases pulmonary dendritic cells and impacts antiviral immune responsiveness. Am J Respir Cell Mol Biol. 2004;30:202–211. doi: 10.1165/rcmb.2003-0259OC. [DOI] [PubMed] [Google Scholar]
  • 86.Moszczynski P, Zabinski Z, Moszczynski P, Jr, Rutowski J, Slowinski S, Tabarowski Z. Immunological findings in cigarette smokers. Toxicol Lett. 2001;118:121–127. doi: 10.1016/s0378-4274(00)00270-8. [DOI] [PubMed] [Google Scholar]
  • 87.Burton RC. Smoking, immunity, and cancer. Med J Aust. 1983;2:411–412. doi: 10.5694/j.1326-5377.1983.tb122541.x. [DOI] [PubMed] [Google Scholar]
  • 88.Bermudez EA, Ridker PM. C-reactive protein, statins, and the primary prevention of atherosclerotic cardiovascular disease. Prev Cardiol. 2002;5:42–46. doi: 10.1111/j.1520-037x.2002.1032.x. [DOI] [PubMed] [Google Scholar]
  • 89.Tracy RP, Psaty BM, Macy E, et al. Lifetime smoking exposure affects the association of C-reactive protein with cardiovascular disease risk factors and subclinical disease in healthy elderly subjects. Arterioscler Thromb Vasc Biol. 1997;17:2167–2176. doi: 10.1161/01.atv.17.10.2167. [DOI] [PubMed] [Google Scholar]
  • 90.Tuomi T, Heliovaara M, Palosuo T, Aho K. Smoking, lung function, and rheumatoid factors. Annals of the rheumatic diseases. 1990;49:753–756. doi: 10.1136/ard.49.10.753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Abhishek A, Butt S, Gadsby K, Zhang W, Deighton CM. Anti-TNF-alpha agents are less effective for the treatment of rheumatoid arthritis in current smokers. J Clin Rheumatol. 2010;16:15–18. doi: 10.1097/RHU.0b013e3181ca4a2a. [DOI] [PubMed] [Google Scholar]
  • 92.Saevarsdottir S, Wedren S, Seddighzadeh M, et al. Patients with early rheumatoid arthritis who smoke are less likely to respond to treatment with methotrexate and tumor necrosis factor inhibitors: observations from the Epidemiological Investigation of Rheumatoid Arthritis and the Swedish Rheumatology Register cohorts. Arthritis and rheumatism. 2011;63:26–36. doi: 10.1002/art.27758. [DOI] [PubMed] [Google Scholar]
  • 93.Sverdrup B, Kallberg H, Bengtsson C, et al. Association between occupational exposure to mineral oil and rheumatoid arthritis: results from the Swedish EIRA case-control study. Arthritis research & therapy. 2005;7:R1296–R1303. doi: 10.1186/ar1824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kleinau S, Erlandsson H, Holmdahl R, Klareskog L. Adjuvant oils induce arthritis in the DA rat. I. Characterization of the disease and evidence for an immunological involvement. J Autoimmun. 1991;4:871–880. doi: 10.1016/0896-8411(91)90050-M. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Cannon GW, Woods ML, Clayton F, Griffiths MM. Induction of arthritis in DA rats by incomplete Freund's adjuvant. J Rheumatol. 1993;20:7–11. [PubMed] [Google Scholar]
  • 96.Lundberg I, Alfredsson L, Plato N, Sverdrup B, Klareskog L, Kleinau S. Occupation, occupational exposure to chemicals and rheumatological disease. A register based cohort study. Scandinavian journal of rheumatology. 1994;23:305–310. doi: 10.3109/03009749409099278. [DOI] [PubMed] [Google Scholar]
  • 97.Linos A, Worthington JW, O'Fallon WM, Kurland LT. The epidemiology of rheumatoid arthritis in Rochester, Minnesota: a study of incidence, prevalence, and mortality. Am J Epidemiol. 1980;111:87–98. doi: 10.1093/oxfordjournals.aje.a112878. [DOI] [PubMed] [Google Scholar]
  • 98.Symmons DP, Barrett EM, Bankhead CR, Scott DG, Silman AJ. The incidence of rheumatoid arthritis in the United Kingdom: results from the Norfolk Arthritis Register. Br J Rheumatol. 1994;33:735–739. doi: 10.1093/rheumatology/33.8.735. [DOI] [PubMed] [Google Scholar]
  • 99.Goemaere S, Ackerman C, Goethals K, et al. Onset of symptoms of rheumatoid arthritis in relation to age, sex and menopausal transition. JRheumatol. 1990;17:1620–1622. [PubMed] [Google Scholar]
  • 100.Silman A, Kay A, Brennan P. Timing of pregnancy in relation to the onset of rheumatoid arthritis. Arthritis Rheum. 1992;35:152–155. doi: 10.1002/art.1780350205. [DOI] [PubMed] [Google Scholar]
  • 101.Wilder RL. Adrenal and gonadal steroid hormone deficiency in the pathogenesis of rheumatoid arthritis. JRheumatolSuppl. 1996;44:10–12. 10–12. [PubMed] [Google Scholar]
  • 102.Wilder RL, Elenkov IJ. Hormonal regulation of tumor necrosis factor-alpha, interleukin-12 and interleukin-10 production by activated macrophages. A disease-modifying mechanism in rheumatoid arthritis and systemic lupus erythematosus? Annals of the New York Academy of Sciences. 1999;876:14–31. doi: 10.1111/j.1749-6632.1999.tb07619.x. [Review] [152 refs] [DOI] [PubMed] [Google Scholar]
  • 103.Allebeck P, Ahlbom A, Ljungstrom K, Allander E. Do oral contraceptives reduce the incidence of rheumatoid arthritis? A pilot study using the Stockholm County medical information system. Scandinavian journal of rheumatology. 1984;13:140–146. doi: 10.3109/03009748409100377. [DOI] [PubMed] [Google Scholar]
  • 104.Linos A, Worthington JW, O'Fallon WM, Kurland LT. Case-control study of rheumatoid arthritis and prior use of oral contraceptives. Lancet. 1983;1:1299–1300. doi: 10.1016/s0140-6736(83)92413-3. [DOI] [PubMed] [Google Scholar]
  • 105.Del Junco DJ, Annegers JF, Luthra HS, Coulam CB, Kurland LT. Do oral contraceptives prevent rheumatoid arthritis? Jama. 1985;254:1938–1941. [PubMed] [Google Scholar]
  • 106.Hannaford PC, Kay CR, Hirsch S. Oral contraceptives and rheumatoid arthritis: new data from the Royal College of General Practitioners' oral contraception study. AnnRheumDis. 1990;49:744–746. doi: 10.1136/ard.49.10.744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Deighton CM, Sykes H, Walker DJ. Rheumatoid arthritis, HLA identity, and age at menarche. AnnRheumDis. 1993;52:322–326. doi: 10.1136/ard.52.5.322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Brun JG, Nilssen S, Kvale G. Breast feeding, other reproductive factors and rheumatoid arthritis. A prospective study. BrJRheumatol. 1995;34:542–546. doi: 10.1093/rheumatology/34.6.542. [DOI] [PubMed] [Google Scholar]
  • 109.Dugowson CE, Koepsell TD, Voigt LF, Bley L, Nelson JL, Daling JR. Rheumatoid arthritis in women. Incidence rates in group health cooperative, Seattle, Washington, 1987–1989. Arthritis Rheum. 1991;34:1502–1507. doi: 10.1002/art.1780341205. [DOI] [PubMed] [Google Scholar]
  • 110.Doran MF, Pond GR, Crowson CS, O'Fallon WM, Gabriel SE. Trends in incidence and mortality in rheumatoid arthritis in Rochester, Minnesota, over a forty-year period. Arthritis and rheumatism. 2002;46:625–631. doi: 10.1002/art.509. [DOI] [PubMed] [Google Scholar]
  • 111.van de Sande MG, de Hair MJ, van der Leij C, et al. Different stages of rheumatoid arthritis: features of the synovium in the preclinical phase. Annals of the rheumatic diseases. 2011;70:772–777. doi: 10.1136/ard.2010.139527. [DOI] [PubMed] [Google Scholar]
  • 112.Lundberg K, Kinloch A, Fisher BA, et al. Antibodies to citrullinated alpha-enolase peptide 1 are specific for rheumatoid arthritis and cross-react with bacterial enolase. Arthritis and rheumatism. 2008;58:3009–3019. doi: 10.1002/art.23936. [DOI] [PubMed] [Google Scholar]
  • 113.Lundberg K, Wegner N, Yucel-Lindberg T, Venables PJ. Periodontitis in RA-the citrullinated enolase connection. Nature reviews. 2010;6:727–730. doi: 10.1038/nrrheum.2010.139. [DOI] [PubMed] [Google Scholar]
  • 114.Mikuls TR. Help stop tooth decay…and prevent RA? J Rheumatol. 2010;37:1083–1085. doi: 10.3899/jrheum.100173. [DOI] [PubMed] [Google Scholar]
  • 115.Mikuls TR, Payne JB, Reinhardt RA, et al. Antibody responses to Porphyromonas gingivalis (P. gingivalis) in subjects with rheumatoid arthritis and periodontitis. International immunopharmacology. 2009;9:38–42. doi: 10.1016/j.intimp.2008.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Wegner N, Wait R, Sroka A, et al. Peptidylarginine deiminase from Porphyromonas gingivalis citrullinates human fibrinogen and alpha-enolase: implications for autoimmunity in rheumatoid arthritis. Arthritis and rheumatism. 2010;62:2662–2672. doi: 10.1002/art.27552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Ramirez AS, Rosas A, Hernandez-Beriain JA, et al. Relationship between rheumatoid arthritis and Mycoplasma pneumoniae: a case-control study. Rheumatology (Oxford) 2005;44:912–914. doi: 10.1093/rheumatology/keh630. [DOI] [PubMed] [Google Scholar]
  • 118.Silman AJ, Pearson JE. Epidemiology and genetics of rheumatoid arthritis. Arthritis research. 2002;4(Suppl 3):S265–S272. doi: 10.1186/ar578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Demoruelle MK, Weisman MH, Simonian PL, et al. Airways abnormalities and rheumatoid arthritis-related autoantibodies in subjects without arthritis: Early injury or initiating site of autoimmunity? Arthritis and rheumatism. 2011 doi: 10.1002/art.34344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Gizinski AM, Mascolo M, Loucks JL, et al. Rheumatoid arthritis (RA)-specific autoantibodies in patients with interstitial lung disease and absence of clinically apparent articular RA. Clin Rheumatol. 2009;28:611–613. doi: 10.1007/s10067-009-1128-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Scher JU, Abramson SB. The microbiome and rheumatoid arthritis. Nature reviews. 2011 doi: 10.1038/nrrheum.2011.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Blankenberg-Sprenkels SH, Fielder M, Feltkamp TE, Tiwana H, Wilson C, Ebringer A. Antibodies to Klebsiella pneumoniae in Dutch patients with ankylosing spondylitis and acute anterior uveitis and to Proteus mirabilis in rheumatoid arthritis. J Rheumatol. 1998;25:743–747. [PubMed] [Google Scholar]
  • 123.Ebringer A, Rashid T, Wilson C. Rheumatoid arthritis, Proteus, anti-CCP antibodies and Karl Popper. Autoimmunity reviews. 2010;9:216–223. doi: 10.1016/j.autrev.2009.10.006. [DOI] [PubMed] [Google Scholar]
  • 124.Vaahtovuo J, Toivanen P, Eerola E. Bacterial composition of murine fecal microflora is indigenous and genetically guided. FEMS microbiology ecology. 2003;44:131–136. doi: 10.1016/S0168-6496(02)00460-9. [DOI] [PubMed] [Google Scholar]
  • 125.Edwards CJ. Commensal gut bacteria and the etiopathogenesis of rheumatoid arthritis. J Rheumatol. 2008;35:1477–14797. [PubMed] [Google Scholar]
  • 126.Linos A, Kaklamani VG, Kaklamani E, et al. Dietary factors in relation to rheumatoid arthritis: a role for olive oil and cooked vegetables? Am J Clin Nutr. 1999;70:1077–1082. doi: 10.1093/ajcn/70.6.1077. [DOI] [PubMed] [Google Scholar]
  • 127.Linos A, Kaklamanis E, Kontomerkos A, et al. The effect of olive oil and fish consumption on rheumatoid arthritis--a case control study. Scandinavian journal of rheumatology. 1991;20:419–426. doi: 10.3109/03009749109096821. [DOI] [PubMed] [Google Scholar]
  • 128.Shapiro JA, Koepsell TD, Voigt LF, Dugowson CE, Kestin M, Nelson JL. Diet and rheumatoid arthritis in women: a possible protective effect of fish consumption. Epidemiology. 1996;7:256–263. doi: 10.1097/00001648-199605000-00007. [DOI] [PubMed] [Google Scholar]
  • 129.Cerhan JR, Saag KG, Merlino LA, Mikuls TR, Criswell LA. Antioxidant micronutrients and risk of rheumatoid arthritis in a cohort of older women. Am J Epidemiol. 2003;157:345–354. doi: 10.1093/aje/kwf205. [DOI] [PubMed] [Google Scholar]
  • 130.Pattison DJ, Silman AJ, Goodson NJ, et al. Vitamin C and the risk of developing inflammatory polyarthritis: prospective nested case-control study. Annals of the rheumatic diseases. 2004;63:843–847. doi: 10.1136/ard.2003.016097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Pattison DJ, Symmons DP, Lunt M, et al. Dietary beta-cryptoxanthin and inflammatory polyarthritis: results from a population-based prospective study. Am J Clin Nutr. 2005;82:451–455. doi: 10.1093/ajcn.82.2.451. [DOI] [PubMed] [Google Scholar]
  • 132.Pattison DJ, Symmons DP, Lunt M, et al. Dietary risk factors for the development of inflammatory polyarthritis: evidence for a role of high level of red meat consumption. Arthritis and rheumatism. 2004;50:3804–3812. doi: 10.1002/art.20731. [DOI] [PubMed] [Google Scholar]
  • 133.Pedersen M, Stripp C, Klarlund M, Olsen SF, Tjonneland AM, Frisch M. Diet and risk of rheumatoid arthritis in a prospective cohort. J Rheumatol. 2005;32:1249–1252. [PubMed] [Google Scholar]
  • 134.Benito-Garcia E, Feskanich D, Hu FB, Mandl LA, Karlson EW. Protein, iron, and meat consumption and risk for rheumatoid arthritis: a prospective cohort study. Arthritis research & therapy. 2007;9:R16. doi: 10.1186/ar2123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Karlson EW, Shadick NA, Cook NR, Buring JE, Lee IM. Vitamin E in the primary prevention of rheumatoid arthritis: the Women's Health Study. Arthritis and rheumatism. 2008;59:1589–1595. doi: 10.1002/art.24194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Costenbader KH, Feskanich D, Holmes M, Karlson EW, Benito-Garcia E. Vitamin D intake and risks of systemic lupus erythematosus and rheumatoid arthritis in women. Annals of the rheumatic diseases. 2008;67:530–535. doi: 10.1136/ard.2007.072736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Costenbader KH, Kang JH, Karlson EW. Antioxidant intake and risks of rheumatoid arthritis and systemic lupus erythematosus in women. Am J Epidemiol. 2010;172:205–216. doi: 10.1093/aje/kwq089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Munger KL, Levin LI, Hollis BW, Howard NS, Ascherio A. Serum 25-hydroxyvitamin D levels and risk of multiple sclerosis. Jama. 2006;296:2832–2838. doi: 10.1001/jama.296.23.2832. [DOI] [PubMed] [Google Scholar]
  • 139.Hernan MA, Olek MJ, Ascherio A. Geographic variation of MS incidence in two prospective studies of US women. Neurology. 1999;53:1711–1718. doi: 10.1212/wnl.53.8.1711. [DOI] [PubMed] [Google Scholar]
  • 140.Arnson Y, Amital H, Shoenfeld Y. Vitamin D and autoimmunity: new aetiological and therapeutic considerations. Annals of the rheumatic diseases. 2007;66:1137–1142. doi: 10.1136/ard.2007.069831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Kamen DL, Cooper GS, Bouali H, Shaftman SR, Hollis BW, Gilkeson GS. Vitamin D deficiency in systemic lupus erythematosus. Autoimmunity reviews. 2006;5:114–117. doi: 10.1016/j.autrev.2005.05.009. [DOI] [PubMed] [Google Scholar]
  • 142.Canning MO, Grotenhuis K, de Wit H, Ruwhof C, Drexhage HA. 1-alpha,25-dihydroxyvitamin D3 (1,25(OH)(2)D(3)) hampers the maturation of fully active immature dendritic cells from monocytes. Eur J Endocrinol. 2001;145:351–357. doi: 10.1530/eje.0.1450351. [DOI] [PubMed] [Google Scholar]
  • 143.Cantorna MT, Hayes CE, DeLuca HF. 1,25-Dihydroxycholecalciferol inhibits the progression of arthritis in murine models of human arthritis. J Nutr. 1998;128:68–72. doi: 10.1093/jn/128.1.68. [DOI] [PubMed] [Google Scholar]
  • 144.Feser M, Derber LA, Deane KD, et al. Plasma 25,OH vitamin D concentrations are not associated with rheumatoid arthritis (RA)-related autoantibodies in individuals at elevated risk for RA. J Rheumatol. 2009;36:943–946. doi: 10.3899/jrheum.080764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Patel S, Farragher T, Berry J, Bunn D, Silman A, Symmons D. Association between serum vitamin D metabolite levels and disease activity in patients with early inflammatory polyarthritis. Arthritis and rheumatism. 2007;56:2143–2149. doi: 10.1002/art.22722. [DOI] [PubMed] [Google Scholar]
  • 146.Andjelkovic Z, Vojinovic J, Pejnovic N, et al. Disease modifying and immunomodulatory effects of high dose 1 alpha (OH) D3 in rheumatoid arthritis patients. Clinical and experimental rheumatology. 1999;17:453–456. [PubMed] [Google Scholar]
  • 147.James MJ, Gibson RA, Cleland LG. Dietary polyunsaturated fatty acids and inflammatory mediator production. Am J Clin Nutr. 2000;71:343S–348S. doi: 10.1093/ajcn/71.1.343s. [DOI] [PubMed] [Google Scholar]
  • 148.Rosell M, Wesley AM, Rydin K, Klareskog L, Alfredsson L. Dietary fish and fish oil and the risk of rheumatoid arthritis. Epidemiology. 2009;20:896–901. doi: 10.1097/EDE.0b013e3181b5f0ce. [DOI] [PubMed] [Google Scholar]
  • 149.Stamp LK, James MJ, Cleland LG. Diet and rheumatoid arthritis: a review of the literature. Semin Arthritis Rheum. 2005;35:77–94. doi: 10.1016/j.semarthrit.2005.05.001. [DOI] [PubMed] [Google Scholar]
  • 150.Simopoulos AP. Omega-3 fatty acids in inflammation and autoimmune diseases. Journal of the American College of Nutrition. 2002;21:495–505. doi: 10.1080/07315724.2002.10719248. [DOI] [PubMed] [Google Scholar]
  • 151.Heliovaara M, Knekt P, Aho K, Aaran RK, Alfthan G, Aromaa A. Serum antioxidants and risk of rheumatoid arthritis. Annals of the rheumatic diseases. 1994;53:51–53. doi: 10.1136/ard.53.1.51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Knekt P, Heliovaara M, Aho K, Alfthan G, Marniemi J, Aromaa A. Serum selenium, serum alpha-tocopherol, and the risk of rheumatoid arthritis. Epidemiology. 2000;11:402–405. doi: 10.1097/00001648-200007000-00007. [DOI] [PubMed] [Google Scholar]
  • 153.Comstock GW, Burke AE, Hoffman SC, et al. Serum concentrations of alpha tocopherol, beta carotene, and retinol preceding the diagnosis of rheumatoid arthritis and systemic lupus erythematosus. Annals of the rheumatic diseases. 1997;56:323–325. doi: 10.1136/ard.56.5.323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Nissen MJ, Gabay C, Scherer A, Finckh A. The effect of alcohol on radiographic progression in rheumatoid arthritis. Arthritis and rheumatism. 62:1265–1272. doi: 10.1002/art.27388. [DOI] [PubMed] [Google Scholar]
  • 155.Maxwell JR, Gowers IR, Moore DJ, Wilson AG. Alcohol consumption is inversely associated with risk and severity of rheumatoid arthritis. Rheumatology (Oxford) 2010;49:2140–2146. doi: 10.1093/rheumatology/keq202. [DOI] [PubMed] [Google Scholar]
  • 156.Cerhan JR, Saag KG, Criswell LA, Merlino LA, Mikuls TR. Blood transfusion, alcohol use, and anthropometric risk factors for rheumatoid arthritis in older women. J Rheumatol. 2002;29:246–254. [PubMed] [Google Scholar]
  • 157.Lu B, Solomon DH, Costenbader KH, Keenan BT, Chibnik LB, Karlson EW. Alcohol consumption and markers of inflammation in women with preclinical rheumatoid arthritis. Arthritis and rheumatism. 2011;62:3554–3559. doi: 10.1002/art.27739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Imhof A, Froehlich M, Brenner H, Boeing H, Pepys MB, Koenig W. Effect of alcohol consumption on systemic markers of inflammation. Lancet. 2001;357:763–767. doi: 10.1016/S0140-6736(00)04170-2. [DOI] [PubMed] [Google Scholar]
  • 159.Cooper CL, Cameron DW. Effect of alcohol use and highly active antiretroviral therapy on plasma levels of hepatitis C virus (HCV) in patients coinfected with HIV and HCV. Clin Infect Dis. 2005;41(Suppl 1):S105–S109. doi: 10.1086/429506. [DOI] [PubMed] [Google Scholar]
  • 160.Boe DM, Nelson S, Zhang P, Quinton L, Bagby GJ. Alcohol-induced suppression of lung chemokine production and the host defense response to Streptococcus pneumoniae. Alcoholism, clinical and experimental research. 2003;27:1838–1845. doi: 10.1097/01.ALC.0000095634.82310.53. [DOI] [PubMed] [Google Scholar]
  • 161.Nelson S, Bagby GJ, Bainton BG, Summer WR. The effects of acute and chronic alcoholism on tumor necrosis factor and the inflammatory response. The Journal of infectious diseases. 1989;160:422–429. doi: 10.1093/infdis/160.3.422. [DOI] [PubMed] [Google Scholar]
  • 162.Szabo G, Mandrekar P, Catalano D. Inhibition of superantigen-induced T cell proliferation and monocyte IL-1 beta, TNF-alpha, and IL-6 production by acute ethanol treatment. Journal of leukocyte biology. 1995;58:342–350. doi: 10.1002/jlb.58.3.342. [DOI] [PubMed] [Google Scholar]
  • 163.Thould AK, Thould BT. Arthritis in Roman Britain. Br Med J (Clin Res Ed) 1983;287:1909–1911. doi: 10.1136/bmj.287.6409.1909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Rothschild BM, Turner KR, DeLuca MA. Symmetrical erosive peripheral polyarthritis in the Late Archaic Period of Alabama. Science. 1988;241:1498–1501. doi: 10.1126/science.3047874. [DOI] [PubMed] [Google Scholar]
  • 165.Fischer KM. Hypothesis: tobacco use is a risk factor in rheumatoid arthritis. Med Hypotheses. 1991;34:116–117. doi: 10.1016/0306-9877(91)90177-z. [DOI] [PubMed] [Google Scholar]
  • 166.Bengtsson C, Nordmark B, Klareskog L, Lundberg I, Alfredsson L. Socioeconomic status and the risk of developing rheumatoid arthritis: results from the Swedish EIRA study. Annals of the rheumatic diseases. 2005;64:1588–1594. doi: 10.1136/ard.2004.031666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Neidell MJ. Air pollution, health, and socio-economic status: the effect of outdoor air quality on childhood asthma. J Health Econ. 2004;23:1209–1236. doi: 10.1016/j.jhealeco.2004.05.002. [DOI] [PubMed] [Google Scholar]
  • 168.Apelberg BJ, Buckley TJ, White RH. Socioeconomic and racial disparities in cancer risk from air toxics in Maryland. Environmental health perspectives. 2005;113:693–699. doi: 10.1289/ehp.7609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Penard-Morand C, Charpin D, Raherison C, et al. Long-term exposure to background air pollution related to respiratory and allergic health in schoolchildren. Clin Exp Allergy. 2005;35:1279–1287. doi: 10.1111/j.1365-2222.2005.02336.x. [DOI] [PubMed] [Google Scholar]
  • 170.Sunyer J. Urban air pollution and chronic obstructive pulmonary disease: a review. Eur Respir J. 2001;17:1024–1033. doi: 10.1183/09031936.01.17510240. [DOI] [PubMed] [Google Scholar]
  • 171.Karakatsani A, Andreadaki S, Katsouyanni K, et al. Air pollution in relation to manifestations of chronic pulmonary disease: a nested case-control study in Athens, Greece. Eur J Epidemiol. 2003;18:45–53. doi: 10.1023/a:1022576028603. [DOI] [PubMed] [Google Scholar]
  • 172.Dockery DW, Pope CA, 3rd, Xu X, et al. An association between air pollution and mortality in six U.S. cities. N Engl J Med. 1993;329:1753–1759. doi: 10.1056/NEJM199312093292401. [DOI] [PubMed] [Google Scholar]
  • 173.van Eeden SF, Yeung A, Quinlam K, Hogg JC. Systemic response to ambient particulate matter: relevance to chronic obstructive pulmonary disease. Proc Am Thorac Soc. 2005;2:61–67. doi: 10.1513/pats.200406-035MS. [DOI] [PubMed] [Google Scholar]
  • 174.Kunzli N, Jerrett M, Mack WJ, et al. Ambient air pollution and atherosclerosis in Los Angeles. Environmental health perspectives. 2005;113:201–206. doi: 10.1289/ehp.7523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Pope CA, 3rd, Burnett RT, Thun MJ, et al. Lung cancer, cardiopulmonary mortality, and long-term exposure to fine particulate air pollution. Jama. 2002;287:1132–41. doi: 10.1001/jama.287.9.1132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Nafstad P, Haheim LL, Oftedal B, et al. Lung cancer and air pollution: a 27 year follow up of 16 209 Norwegian men. Thorax. 2003;58:1071–1076. doi: 10.1136/thorax.58.12.1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Pereira FA, de Assuncao JV, Saldiva PH, Pereira LA, Mirra AP, Braga AL. Influence of air pollution on the incidence of respiratory tract neoplasm. J Air Waste Manag Assoc. 2005;55:83–87. doi: 10.1080/10473289.2005.10464603. [DOI] [PubMed] [Google Scholar]
  • 178.Schwartz J, Dockery DW, Neas LM. Is daily mortality associated specifically with fine particles? J Air Waste Manag Assoc. 1996;46:927–939. [PubMed] [Google Scholar]
  • 179.Schwartz J, Laden F, Zanobetti A. The concentration-response relation between PM(2.5) and daily deaths. Environmental health perspectives. 2002;110:1025–1029. doi: 10.1289/ehp.021101025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Laden F, Neas LM, Dockery DW, Schwartz J. Association of fine particulate matter from different sources with daily mortality in six U.S. cities. Environmental health perspectives. 2000;108:941–947. doi: 10.1289/ehp.00108941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Costenbader KH, Chang SC, Laden F, Puett R, Karlson EW. Geographic variation in rheumatoid arthritis incidence among women in the United States. Archives of internal medicine. 2008;168:1664–1670. doi: 10.1001/archinte.168.15.1664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Vieira VM, Hart JE, Webster TF, et al. Association between residences in U.S. northern latitudes and rheumatoid arthritis: A spatial analysis of the Nurses' Health Study. Environmental health perspectives. 2010;118:957–961. doi: 10.1289/ehp.0901861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Silman AJ, MacGregor AJ, Thomson W, et al. Twin concordance rates for rheumatoid arthritis: results from a nationwide study. BrJRheumatol. 1993;32:903–907. doi: 10.1093/rheumatology/32.10.903. [DOI] [PubMed] [Google Scholar]
  • 184.Linn-Rasker SP, van der Helm-van Mil AH, van Gaalen FA, et al. Smoking is a risk factor for anti-CCP antibodies only in rheumatoid arthritis patients who carry HLA-DRB1 shared epitope alleles. Annals of the rheumatic diseases. 2006;65:366–371. doi: 10.1136/ard.2005.041079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Lee HS, Irigoyen P, Kern M, et al. Interaction between smoking, the shared epitope, and anti-cyclic citrullinated peptide: a mixed picture in three large North American rheumatoid arthritis cohorts. Arthritis and rheumatism. 2007;56:1745–1753. doi: 10.1002/art.22703. [DOI] [PubMed] [Google Scholar]
  • 186.Keenan BT, Chibnik LB, Cui J, et al. Effect of interactions of glutathione S-transferase T1, M1, and P1 and HMOX1 gene promoter polymorphisms with heavy smoking on the risk of rheumatoid arthritis. Arthritis and rheumatism. 2010;62:3196–3210. doi: 10.1002/art.27639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Criswell LA, Saag KG, Mikuls TR, et al. Smoking interacts with genetic risk factors in the development of rheumatoid arthritis among older Caucasian women. Annals of the rheumatic diseases. 2006;65:1163–1167. doi: 10.1136/ard.2005.049676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Mikuls TR, Gould KA, Bynote KK, et al. Anticitrullinated protein antibody (ACPA) in rheumatoid arthritis: influence of an interaction between HLA-DRB1 shared epitope and a deletion polymorphism in glutathione S-transferase in a cross-sectional study. Arthritis research & therapy. 2010;12:R213. doi: 10.1186/ar3190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Mikuls TR, Levan T, Gould KA, et al. Impact of interactions of cigarette smoking with NAT2 polymorphisms on rheumatoid arthritis risk in African Americans. Arthritis and rheumatism. 2012;64:655–664. doi: 10.1002/art.33408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Hill JA, Southwood S, Sette A, Jevnikar AM, Bell DA, Cairns E. Cutting edge: the conversion of arginine to citrulline allows for a high-affinity peptide interaction with the rheumatoid arthritis-associated HLA-DRB1*0401 MHC class II molecule. J Immunol. 2003;171:538–541. doi: 10.4049/jimmunol.171.2.538. [DOI] [PubMed] [Google Scholar]
  • 191.Karlson EW, Chibnik LB, Kraft P, et al. Cumulative association of 22 genetic variants with seropositive rheumatoid arthritis risk. Annals of the rheumatic diseases. 2010 doi: 10.1136/ard.2009.120170. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Demoruelle MK, Deane K. Antibodies to Citrullinated Protein Antigens (ACPAs): Clinical and Pathophysiologic Significance. Curr Rheumatol Rep. 2011 doi: 10.1007/s11926-011-0193-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Cooles FA, Isaacs JD. Pathophysiology of rheumatoid arthritis. Current opinion in rheumatology. 2011;23:233–240. doi: 10.1097/BOR.0b013e32834518a3. [DOI] [PubMed] [Google Scholar]
  • 194.Kakumanu P, Sobel ES, Narain S, et al. Citrulline dependence of anti-cyclic citrullinated peptide antibodies in systemic lupus erythematosus as a marker of deforming/erosive arthritis. J Rheumatol. 2009;36:2682–2690. doi: 10.3899/jrheum.090338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Alenius GM, Berglin E, Rantapaa Dahlqvist S. Antibodies against cyclic citrullinated peptide (CCP) in psoriatic patients with or without joint inflammation. Annals of the rheumatic diseases. 2006;65:398–400. doi: 10.1136/ard.2005.040998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Kuhn KA, Kulik L, Tomooka B, et al. Antibodies against citrullinated proteins enhance tissue injury in experimental autoimmune arthritis. J Clin Invest. 2006;116:961–973. doi: 10.1172/JCI25422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Sokolove J, Zhao X, Chandra PE, Robinson WH. Immune complexes containing citrullinated fibrinogen costimulate macrophages via Toll-like receptor 4 and Fcgamma receptor. Arthritis and rheumatism. 2011;63:53–62. doi: 10.1002/art.30081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Zhao X, Okeke NL, Sharpe O, et al. Circulating immune complexes contain citrullinated fibrinogen in rheumatoid arthritis. Arthritis research & therapy. 2008;10:R94. doi: 10.1186/ar2478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Okamoto S, Adachi M, Chujo S, et al. Etiological role of cigarette smoking in rheumatoid arthritis: Nasal exposure to cigarette smoke condensate extracts augments the development of collagen-induced arthritis in mice. Biochemical and biophysical research communications. 2011;404:1088–1092. doi: 10.1016/j.bbrc.2010.12.118. [DOI] [PubMed] [Google Scholar]
  • 200.Lindblad SS, Mydel P, Jonsson IM, Senior RM, Tarkowski A, Bokarewa M. Smoking and nicotine exposure delay development of collagen-induced arthritis in mice. Arthritis research & therapy. 2009;11:R88. doi: 10.1186/ar2728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Breban MA, Moreau MC, Fournier C, Ducluzeau R, Kahn MF. Influence of the bacterial flora on collagen-induced arthritis in susceptible and resistant strains of rats. Clinical and experimental rheumatology. 1993;11:61–64. [PubMed] [Google Scholar]
  • 202.Moudgil KD, Kim E, Yun OJ, Chi HH, Brahn E, Sercarz EE. Environmental modulation of autoimmune arthritis involves the spontaneous microbial induction of T cell responses to regulatory determinants within heat shock protein 65. J Immunol. 2001;166:4237–4243. doi: 10.4049/jimmunol.166.6.4237. [DOI] [PubMed] [Google Scholar]
  • 203.Kinloch AJ, Alzabin S, Brintnell W, et al. Immunization with Porphyromonas gingivalis enolase induces autoimmunity to mammalian alpha-enolase and arthritis in DR4-IE-transgenic mice. Arthritis and rheumatism. 2011;63:3818–3823. doi: 10.1002/art.30639. [DOI] [PubMed] [Google Scholar]

RESOURCES