Journal Information
Vol. 8. Issue 6.
Pages 342-350 (November - December 2012)
Visits
14695
Vol. 8. Issue 6.
Pages 342-350 (November - December 2012)
Review Article
Full text access
Tobacco and Other Environmental Risk Factors in Rheumatoid Arthritis
Tabaco y otros factores ambientales en la artritis reumatoide
Visits
14695
Virginia Ruiz-Esquide
Corresponding author
vruizesquide@yahoo.com

Corresponding author.
, Raimon Sanmartí
Unidad de Artritis, Servicio de Reumatologia, Hospital Clínic de Barcelona, Barcelona, Spain
This item has received
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Tables (3)
Table 1. Epidemiological Studies on the Risk of Rheumatoid Arthritis and Smoking.
Table 2. Studies on Relative Risk of Developing RA in Relation to Tobacco Use and HLADRB Genotype (Rheumatoid Epitope).
Table 3. Effect of Tobacco on Treatment Response.
Show moreShow less
Abstract

Many environmental factors have been associated with an increased risk of developing Rheumatoid Arthritis (RA), but so far smoking is the only environmental risk factor that has been extensively studied and widely accepted. Smoking is associated with an increased risk of developing seropositive RA (RF and/or ACPA). Recent studies show that tobacco smoking can influence disease phenotype, with the development of more aggressive disease and greater joint damage; but other studies show contradictory results. Recent data suggests that response to antirheumatic therapy in RA is worse in smokers. In this article we review different environmental factors that have been associated with an increased risk of developing RA, with a special interest in tobacco smoking.

Keywords:
Tobacco
Rheumatoid arthritis
Environmental risk factors
Resumen

Existen distintos factores ambientales implicados en la patogenia de la artritis reumatoide, aunque es el tabaco el factor más ampliamente estudiado y reconocido. El tabaquismo está asociado a un incremento del riesgo de artritis reumatoide seropositiva (FR y/o ACPA). Además estudios recientes ponen de manifiesto que el consumo de tabaco puede influir en la expresión clínica de la enfermedad, determinar un curso evolutivo más grave y una mayor destrucción articular, aunque no todos los estudios son concordantes. Datos recientes sugieren que la respuesta al tratamiento antirreumático sería peor en los enfermos fumadores. En el presente artículo se revisan los distintos factores ambientales que han sido implicados en la AR, con especial énfasis en el tabaquismo.

Palabras clave:
Tabaco
Artritis reumatoide
Factores de riesgo ambientales
Full Text
Introduction

Rheumatoid arthritis (RA) is the most common chronic inflammatory joint disease, affecting about 0.5%–1% of the general population and causing progressive joint destruction, disability and reduced life expectancy. The etiology of RA is unknown and its pathogenesis is only partially explained to date. In recent years we have studied and identified multiple risk factors for its development. We know that genetic and environmental factors are involved and their interaction may be decisive in the development of the disease. Among the environmental factors is tobacco, which has been extensively studied and is now recognized as an important risk factor for RA.1

Rheumatoid Arthritis and Genetic Risk Factors

For over 30 years we have known that there is a genetic susceptibility factor in RA2 which contributes 50%–60% to the development of the disease.3 Several studies have reported the presence of the disease in between 2% and 4% in dizygotic twins and between 12% and 15% in monozygotic twins.3,4 Thus, those with a first degree relative with RA may have from 2 to 10 times greater risk of developing the disease than the general population.5 Initially, an association of RA with HLADRB1 *04 and some of its alleles (*0401, *0404, *0405 or *0408) was found, and later confirmed with other alleles HLADRB as *0101, *0102 or *010. Today we know that all these alleles encode the same amino acid sequence of the third hypervariable region of the beta chain of the HLA molecule, a region that is central to the process known as antigen recognition, and has been called rheumatoid epitope (RE) or shared epitope (SE). The presence of these alleles not only increases the risk of developing RA, especially seropositive disease (positive rheumatoid factor (RF) and/or antibodies against citrullinated proteins/peptides (ACPA)) but their presence in numerous studies has been associated to environmental factors such as tobbaco,6–10 as well as a worse prognosis, with a greater degree of joint destruction and presence of extra-articular11–13 manifestations. This poor prognosis is particularly evident when the RE is included within the HLA-DRB1 *04, especially with allele *0401. But HLA-DRB1 accounts for approximately one third of the genetic component of RA,14,15 and an intense search has been undertaken for other non-HLA genes associated with RA. In association studies of candidate genes many genetic polymorphisms that also contribute to the development of RA, but to a lesser extent thn the RE, have been identified. Between them, PTPN22 (a functional variant of the protein tyrosine phosphatase intracellular N22) stands out as the second largest susceptibility gene for the development of the RA, which doubles the risk of seropositive RA in heterozygous and four times in homozigotes.16,17 Other RA susceptibility genes in populations of European descent are STAT4,18,19 a key transcription factor in regulating the immune response involved in signaling pathways that promote differentiation of CD4 Th1 and Th,17 which are involved in the pathogenesis of RA and TRAF1/C5.20,21 The recent availability of whole genome studies (GWASs) has allowed the identification of more than 20 susceptibility locus for RA, whose contribution to genetic variation as a whole could be around 5%.20

Rheumatoid Arthritis and Non-genetic Risk Factors

As mentioned, genetic factors warrant about 50% of the risk of RA, leaving the rest to other factors. We have studied various environmental factors, although the scientific evidence on their exact involvement is inconclusive in many cases. We review briefly the most important environmental factors.

Hormones. The higher prevalence of RA in women, especially during childbearing years and frequent disease improvement during pregnancy22 has led to the identification of the possible hormonal role in disease susceptibility. There is considerable controversy as to whether oral contraceptives decrease the risk of developing RA, with some studies finding a clear association,23–25 others,26–30 including a metanalysis31 not showing a lower incidence of RA in women treated with oral contraceptives. One such study noted, however, that early menarche or extended breastfeeding (more than 12 months total) decrease the risk of RA,29 although this last point also is controversial because some studies have observed the opposite effect.24 A recently published study indicates that early menopause favors the risk of developing RA.32 Moreover, in men with RA, male sex hormone levels, especially testosterone, have been found decreased, while no differences were observed in the levels of sex hormones in women with RA and healthy controls.33

Socioeconomic factors. Socioeconomic status influences the course of the disease, but may also determine an increased risk of developing it.34 There was an inverse association between level of formal education and socioeconomic status defined by work activity and the risk of RA.35,36

Dietary factors. It has been suggested that a Mediterranean diet, rich in fish, olive oil, cooked vegetables and fruit has a protective effect against RA which could be due to the high content of omega 3 in such a diet.37,38 Consumption of red meat would have no effect on the risk of developing RA.39

Vitamin D. Vitamin D has been widely studied regarding its involvement in various autoimmune diseases. Its role in relation to risk of developing RA is “misleading”,40,41 although there seems to be an inverse association between consumption of vitamin D and development of RA,42 and between serum levels of vitamin D and disease progression, showing increased activity of the disease and more disability at lower levels of vitamin D.43–45

Alcohol. According to a recently published Danish study, alcohol consumption may have a protective effect on RA, in a dose dependent manner,27 observing that the magnitude of risk reduction would be greater in smokers and bearers of the SE.46

Coffee. Multiple studies have analyzed the effect of coffee consumption on RA but the results are discordant,47–49 although there could be an increased risk of RA in relation to consumption of high doses of coffee (more than 10 cups a day).27

Infections. Several infectious agents have been studied and implicated in the development of RA based on a higher frequency of positive viral serology or viral presence in synovial fluid of RA patients; however, their role as triggering agents of the disease is still controversial. Perhaps these agents may have some involvement in the development of the disease in the context of genetic predisposition and not in isolation, but together with other interacting risk factors. It should be noted the great interest in recent years Porphyromonas gingivalis has aroused as a potential stimulus for the development of RA. P. gingivalis is the major causative agent of periodontitis, a disease that is more frequent, about double, in subjects with RA than in the healthy population.50 It is the only bacterium known to express the enzyme peptidylarginine-deiminase (PAD), responsible for citrullination of proteins,51 and produces chronic inflammation (characterized by the presence of proinflammatory cytokines and TNF) and erosive destruction of periodontal bone.52 Like RA, periodontal disease has been associated with HLA-DRB04.53

Silica. Exposure to crystalline silica is a well defined risk factor for RA. Silica is present in the mining, construction, ceramics, glass, as well as in the agriculture sector and in electronics, and doubles the risk of RA in an analysis adjusting for exposure to tobacco.54

Tobacco. Tobacco is the most widely studied and recognized environmental risk factor for development of RA. Over 20 years ago Vassey et al.30 first suggested its involvement in RA, unexpectedly observing this effect in their study on the effect of oral contraceptives in RA. Since then this effect of tobacco has been reproduced and confirmed in multiple studies and case-control cohorts.1,55–58 These studies examined the effect of tobacco on RA as a risk factor for its development and its strong interaction with genetic factors and ACPA, as well as its effect on clinical, radiological and response to disease modifying treatments, leading to a better understanding of the disease. We discuss in detail the effect of tobacco in the various aspects of RA.

Rheumatoid Arthritis and TobaccoTobacco and Immunity

The use of tobacco affects multiple organs including the respiratory and cardiovascular systems, but also affects the immune system by producing an inflammatory response. It has been observed that tobacco affects both the humoral and cellular immune response and could have both pro-and anti-inflammatory effects as immunosuppressants through diverse59 mechanisms. On one side it has been described that it may increase the inflammatory response observed in smokers by increasing serum fibrinogen, the activity of autoreactive B cells and an increase in acute phase reactants and pro-inflammatory cytokines such as TNF-alpha, IL6 as well as circulating polymorphonuclear cells. However it is also known for its immunosuppressive effects, such as a reduction of circulating immunoglobulins and inhibition of cytokines such as IL-1B, IL-2 and gamma-interferon or IL-8 release by endothelial cells. It would also have effects on dendritic cells and antigen presentation and inhibition of macrophage function against intracellular microorganisms.60

It has been observed that the use of tobacco modulates cell proliferation and death, inducing new epitopes either directly through the oxidation of existing epitopes or indirectly by interfering with the elimination of apoptotic cells with the consequent exposure of sequestered intracellular antigens by the immune system and stimulating the population of antigen presenting cells present in the lungs, thereby amplifying new antigen presenting capacity,61 facilitating the development of autoimmunity. Furthermore tobacco smoke contains very high concentrations of free radicals and may also increase the generation and activation of endogenous free radicals. These toxins interact with DNA and may cause mutation or gene activation that ultimately can trigger autoimmune phenomena and autoinmune disease.62 Thus, use of tobacco has been causally linked to the development and expression of multiple autoimmune diseases including RA,30,56 systemic lupus erythematosus,63,64 multiple sclerosis,65,66 Graves disease67,68 and primary biliary cirrhosis69 among others.

Tobacco and Risk of Rheumatoid Arthritis

As previously mentioned, after the first description of the association of tobacco with RA in 1987,30 two large prospective studies were then published in the 1990s, the first,56 a cohort study of 121700 nurses in which a slight increased risk of developing RA in relation to non-smokers was observed, with an RR for active smokers of 1.3 (95% CI 0.9–2.1) and RR for ex-smokers of 1.5 (95% CI 0.9–2.3), as well as a second study1 of more than 50000 subjects that showed an increased risk of developing seropositive RA (RF positive) in active smokers (RR 3.8, 95% CI 2.0–6.9) and former smokers (RR 2.6, 95% CI 1.3–5.3) compared with nonsmokers. Many other studies confirmed this association between tobacco and RA10,57,70–72 and several of them noted that the excess risk persisted after quitting smoking for 10 or even 20 years,27,49,73–78 that the probably years of consumption of tobacco weighed more than the number of cigarettes smoked daily in relation to the risk of RA58 and that a dose effect could be seen, i.e. the greater the number of cigarettes per day and pack/years smoked increased the risk of RA.27,74Table 1 summarizes the epidemiological studies on smoking and risk of RA (Sugiyama et al.79 modified).

Table 1.

Epidemiological Studies on the Risk of Rheumatoid Arthritis and Smoking.

Authors/Year  Type of Study  Patients (Cases/Controls or Cohort)  Population Gender  Smokers  RR/OR Active Smokers  Ex-Smokers 
Vessey et al.,30 1987  Cohort  78/600  Women  1.50 (0.93–2.44)  1.66 (1.00–2.78)  1.15 (0.54 –2.43) 
Hernández-Ávila et al.,56 1990  Cohort  217/121.700  Women    1.3 (0.9–2.1)  1.5 (0.9–2.3) 
Heliovaara et al.,1 1993  Cohort  161/28.364  Men  2.04 (1.10–3.79)  1.60 (0.70–3.80)  1.40 (0.50–3.80) 
      RF+  3.86 (2.71–5,49)  4.31 (2.87–6,50)  2.80 (1.40–5,60) 
      RF−  0.81 (0.49–1.34)  0.66 (0.34–1.27)  1.10 (0.50–2.40) 
    351/24.445  Women  1.06 (0.84–1.35)  1.10 (0.84–1.44)  0.92 (0.53–1.58) 
      RF+  1.04 (0.78–1.40)  1.10 (0.80–1.52)  0.80 (0.40–1.70) 
      RF−  1.10 (0.73–1.66)  1.10 (0.70–1.80)  1.10 (0.50–2.60) 
Criswell et al.,73 2002  Cohort  154/31.336  Women  1.70 (1.02–2.85)  2.20 (1.4–2.00)  1.30 (0.80–3.30) 
      >20 p/year  1.99 (1.41–2.81)  –  – 
      <20 p/year  1.10 (0.60–1.80)    – 
Padyukov et al.,10 2004  Cases-controls  858/1.48  Both    1.50 (1.20–2.00)   
      RF+    2.20 (1.70–3.00)   
      RF−    0.80 (0.60–2.20)   
    246/312  Men    1.70 (1.00–2.90)   
      RF+    3.30 (1.70–2.90)   
      RF−    0.60 (0.30–1.30)   
    612/736  Women    1.50 (1.10–2.00)   
      RF+    2.20 (1.60–3.00)   
      RF−    0.80 (0.60–1.30)   
Heliovaara et al.,49 2000  Cohort  7.697/377.481  RF+  1.85 (1.14–2.709  1.93 (1.14–3.28)  1.76 (0.92–3.38) 
Costenbader et al.,74 2006  Cohortes  680/103.818  Women  1.46 (1.24–1.71)  1.43 (1.16–1.75)  1.47 (1.24–1.71) 
      RF+  1.59 (1.29–1.97)  1.58 (1.21–2.06)  1.60 (1.27–2.02) 
      RF−  1.28 (1.00–1.65)  1.23 (0.88–1.70)  1.31 (1.00–1.73) 
      >20 p/year  1.72 (1.41–2.11)  –  – 
      <20 p/year  1.19 (0.95–1.50)  –  – 
Stolt et al.,75 2003  Cases-controls  190/245  Men  1.40 (0.80–2.30)  1.30 (0.71–2.40)  1.40 (0.80–2.50) 
      RF+  1.90 (1.00–3.50)  1.80 (0.80–4,10)  1.90 (0.90–3.80) 
      RF−  0.80 (0.40–1.60)  0.70 (0.30–1.60)  0.90 (0.40–1.90) 
    489/602  Women  1.30 (1.00–1.70)  1.40 (1.00–2.00)  1.20 (0.90–1.70) 
      RF+  1.70 (1.20–2.30)  1.80 (1.30–2.60)  1.60 (1.10–2.30) 
      RF−  0.80 (0.60–1.20)  0.90 (0.50–1.40)  0.80 (0.50–1.30) 
Karlson et al.,58 1999  Cohort    Women  1.13 (1.08–1.18)  1.18 (1.07–1.31)  1.06 (0.96–1.13) 
Pedersen et al.,27 2006  Cases-Controls  515/769  Global  1.70 (1.38–2.10)  1.80 (1.37–2.36)  1.57 (1.13–2.19) 
      CCP+  1.66 (1.23–2.24)  1.73 (1.17–2.56)  1.57 (0.99–2.48) 
      CCP−  1.04 (0.65–1.68)  0.83 (0.49–1.39)  1.35 (0.76–2.39) 
    149/291  Men  1.75 (1.15–2.65)  1.89 (1.09–3.30)  1.58 (0.84–2.97) 
      >20 p/year  2.00 (1.12–3.58)  –  – 
      <20 p/year  1.63 (0.99–2.70)  –  – 
    366/478  Women  1.79 (1.38–2.97)  1.84 (1.33–2.54)  1.69 (1.12–2.55) 
      >20 p/year  2.07 (1.35–3.16)  –  – 
      <20 p/year  1.68 (1.27–2.26)  –  – 
Voigt et al.,76 1994  Cases-controls  349/1.457  Women  1.31 (1.07–1.60)  1.33 (1.00–1.77)  1.28 (0.96–1.71) 
      >20 p/year  1.16 (0.90–1.50)  –  – 
      <20 p/year  1.49 (1.06–2.10)  –  – 
Olsson et al.,78 2004    74/382  Men  2.54 (1.55–4.16)  2.90 (1.40–6,40)  2.30 (1.20–4.40) 
      RF+  4.60 (2.20–9.60)  5,80 (1.90–17.10)  3.80 (1.40–10.20) 
      >20 p/year  2.50 (1.20–5.10)  –  – 
      <20 p/year  2.20 (1.15–4.22)  –  – 
    159/368  Women  1.52 (1.09–2.12)  1.80 (1.10–2.90)  1.30 (0.80–2.90) 
      RF+  1.56 (1.03–2.36)  1.8 (1.00–3.50)  1.40 (0.80–2.40) 
      >20 p/year  1.33 (0.86–2.04)  –  – 
      <20 p/year  1.60 (0.90–3.10)  –  – 

Modified from Sugiyama et al.79

Approximately one third of ACPA positive RA patients could be attributed to the consumption of tobacco. In a recent study, smoking would be responsible for 35% of ACPA positive RA (95% CI 25%–45%), this effect being greater in men than in women (42% and 31%, respectively) and an even greater in the presence of two copies of the rheumatoid epitope (55% CI 95%: 3%–67%).80 This effect is comparable in intensity to that of smoking in coronary artery disease.

Tobacco, Antibodies to Citrullinated Proteins/Peptides/Rheumatoid Factor and HLA-DRB1

It has been recently noted that the use of tobacco is selectively associated with increased risk of seropositive RA (RF and/or ACPA positive). Independent studies have shown that smoking increases the risk of seropositive but not seronegative RA, both in Caucasian (northern Europe)27,75,81–86 as Latinamerican9 populations. However, a study that analyzed an African-American population with RA failed to show this association87 and in one of three large U.S. cohorts, this association was observed exclusively in heavy smokers.88

The first studies examining the relationship between tobacco and RA observed the existence of a higher frequency of RF positivity and higher RF titers in smokers compared to those of non-smokers..8275,89 Later studies also confirmed that ACPA90,91 are more frequent in smokers with RA. It is known that there is a high correlation between the presence of ACPAs and RF, but some authors have analyzed both antibodies in smokers with RA, and a relationship has been seen with ACPA but not with RF.92 In another study where a subanalysis evaluating patients discordant for ACPA and RF was conducted, there were similar proportions of smokers in both groups (ACPA+/FR− vs ACPA−/FR+),90 suggesting an association of tobacco with both autoantibodies.

The increased risk of seropositive RA in smokers is associated with the presence of rheumatoid epitope (HLA-DRB1), showing that there is significant gene–environment interaction between the alleles of the RE and tobacco. This fact is clearly seen in a Swedish cohort of RA10 which analyzed the interaction between RE and smoking. The authors found that while in patients with one and two copies of the RE the relative risk of developing seropositive RA was 2.4 (95% CI 1.4–4.2) and 4.2 (95% CI 2.1–8.3), respectively, compared to those without RE, in smokers the risk increased to 5.5 (95% CI 3.0–10.0) in the presence of a copy of the RE and 15.7 (95% CI 7.2–34.2) in the presence of two copies of the RE. Neither RE nor tobacco nor the combination of both factors were associated with an increased risk of seronegative RA. Later studies found similar results8,92–95 (Table 2).

Table 2.

Studies on Relative Risk of Developing RA in Relation to Tobacco Use and HLADRB Genotype (Rheumatoid Epitope).

Author  Population  Sample Size  Result 
Padyukov et al.,10 2004  Sweden  858/1.048Cases-controls  RF+/ET−/RE−/− RR: 1RF+/ET+/RE−/− RR: 2.4 (1.3–4.6)RF+/ET−/RE−/+ RR: 2.4 (1.4-4.2)RF+/ET+/RE−/+ RR: 5.5 (3.0–10.0)RF+/ET−/RE+/+ RR: 4.2 (2.1–8.3)RF+/ET+/RE+/+ RR: 15.7 (7.2–34.2)RF−/ET−/RE−/− RR: 1RF−/ET+/RE−/− RR: 0.6 (0.6–1.1)RF−/ET−/RE−/+ RR: 0.9 (0.6–1.5)RF−/ET+/RE−/+ RR: 0.9 (0.5–1.7)RF−/ET−/RE+/+ RR: 0.7 (0.4–1.6)RF−/ET+/RE+/+ RR: 1.2 (0.5–3.0) 
Kallberg et al.,8 2007EIRA study  Sweden  1.970/2.405Cases-controls  Anti-CCP+/ET−/RE −/− RR: 1Anti-CCP+/ET+/RE−/− RR: 1.3 (0.7–2.2)Anti-CCP+/ET−/RE−/+ RR: 2.7 (1.6–4.7)Anti-CCP+/ET+/RE−/+ RR: 5.4 (3.3–8.9)Anti-CCP+/ET−/RE+/+ RR: 5.1 (2.7–9.8)Anti-CCP+/ET+/RE+/+ RR: 23.6 (12.8–43.8)Anti-CCP−/ET−/RE−/− RR: 1Anti-CCP−/ET+/RE−/− RR: 0.7 (0.5–1.1)Anti-CCP−/ET−/RE−/+ RR: 1.0 (0.6–1.5)Anti-CCP−/ET+/RE−/+ RR: 1.0 (0.7–1.5)Anti-CCP−/ET−/RE+/+ RR: 0.9 (0.5–1.8)Anti-CCP−/ET+/RE+/+ RR: 1.1 (0.6–2.2) 
Pedersen et al.,89 2007  Denmark  309/445Cases-controls  ET−/RE−/− RR: 1ET+/RE−/+ RR: 1.74 (0.76–3.98)ET−/RE−/+ RR: 3.31 (1.58–6.96)ET+/RE−/+ RR: 5.07 (2.48–10.4)ET−/RE+/+ RR: 17.4 (7.22–41.8)ET+/RE+/+ RR: 57.4 (22.0–150) 
Van der Helm-Van Mil et al.,90 2007  Holland  142/279RA vs undifferentiated arthritis  RE−/Anti-CCP−/ET− RR: 1RE−/Anti-CCP−/ET+ RR: 1 (0.5–2.2)RE−/Anti-CCP+/ET− RR: 7.5 (2.1–28.5)RE−/Anti-CCP+/ET+ RR: 9.4 (2.4–39.8)RE+/Anti-CCP−/ET− RR: 1RE+/Anti-CCP−/ET+ RR: 0.5 (0.2–1.2)RE+/Anti-CCP+/ET− RR: 3.3 (1.3–8.3)RE+/Anti-CCP+/ET+ RR: 8.0 (3.1–21.1) 

RR: relative risk; RF: Rheumatoid Factor; ET: Exposure to tobacco; RE: Rheumatoid epitope.

We hypothesize that the exposure to tobacco and the local inflammatory reaction and cell necrosis produced by it stimulates protein citrullination in the lungs thus providing a substrate for the activation of an immune response. Following this hypothesis we have studied the presence of citrullinated peptides and proteins in the bronchoalveolar lavage (BAL) of smokers noting in the latter that there is a significant increase relative to non-smokers.90 Along the same lines, another study has observed an increased expression of PAD in bronchoalveolar cells of smokers.96 The presence of genetic susceptibility factors for the citrullination of proteins in the lungs could facilitate the development of a local autoimmune response with production of anti-citrullinated peptide antibodies. At a later moment, the presence of articular inflammatory processes and therefore citrullinated peptides97 could form immunocomplexes with the circulating anti-citrulline, eventually triggering an immune response with release of inflammatory mediators (TNF, ILs).98

It is known that anti-citrullinated peptides (ACPA) can be detected in the serum years before the onset of clinical manifestations of RA.99,100 According to the hypothesis above, the prevalence of ACPA in smokers may be higher than in the general population, but this fact has been scarcely studied. Our group observed a similar frequency of ACPA in the serum of heavy smokers without RA and a control population (1.8% vs 1.9%), although in those with COPD, the frequency was slightly higher (3.7%).101 These observations are similar to those observed by Wood et al.102 group.

Tobacco and Clinical Course of Disease

Tobacco not only increases the risk of seropositive RA but also might influence the phenotype or clinical expression of disease.

RA patients who smoke have an earlier onset of disease.83,85,86,103–105 Most studies agree that other baseline characteristics such as duration and disease activity or disability, are comparable to those of non-smoking patients,104–106 although some observed an increased in baseline activity of the disease in smokers.83,84 The impact of tobacco in the clinical course of disease is unclear since some studies report that smokers have a worse outcome with greater disability and disease activity83,85,86,107; others show a similar pattern of disease in smokers and non-smokers.84,103,105 On the other hand, some studies indicate a higher frequency of extra-articular manifestations in smoking RA patients compared to non-smokers,108 mainly higher frequency of rheumatoid nodules82,83,87,103 and lung involvement.109,110

In recent years several studies have examined specifically whether smoking is a factor of poor response to antirheumatic treatment with DMARDs or TNF antagonists. A study shows that smokers used more DMARDs and higher doses than non-smokers,85 while two other studies confirmed that smoking is a factor of poor response to treatment with methotrexate in monotherapy.86,111 This would agree with data recently published from a Swedish registry study that evaluated the influence of tobacco in the response to methotrexate in a cohort of early onset RA; the authors found that after three months of treatment, nonsmokers achieved a significantly higher good EULAR response than smokers (36% vs 27%), with smokers less likely to achieve remission.112 This study also analyzed the response to anti-TNF as a first biologic after DMARD failure in smokers and nonsmokers and found that active smokers were less likely to achieve a good EULAR response at 3 months, succeeding only in 29% of current smokers compared with 43% of nonsmokers. In analyzing the probability of remission in both groups it found that active smokers had a tendency to a lower likelihood of achieving this objective at 3 months, this being significant at 6 months with an OR 0.54 (95% CI 0.30–0.99). Several other studies have analyzed the response to anti-TNF treatment in relation to consumption of tobacco and observed similar results, confirming a lower response to anti-TNF in smoking patients112–115 (Table 3). In addition, a subanalysis of the BeSt study identified several risk factors for reactivation of arthritis in patients who could discontinue infliximab after achieving good disease control, including having the RE, but also smoking.116

Table 3.

Effect of Tobacco on Treatment Response.

Author/Research Group  Study Design  Population  Sample Size  Response to Treatment  Results 
Söderlin et al.,85 2011Data from BARFOT Study  Retrospective  Sweden  1.787  DMARD  Good EULAR response after treatment onset:At 3 months: active smokers 25% vs non-smokers 33% (P=.011)At 6 months: active smokers 26% vs non-smokers 33% (P=.018)At 12 months: active smokers 30% vs non-smokers 37% (P=.013) 
Hyrch et al.,110 2006  Retrospective  United Kingdom  2.879  Anti-TNF  Patients treated with infliximab: significant association between active smoking and less treatment response at 6 months OR 0.77 (95% CI 0.60–0.99)Patients treated with etanercept: no differences in EULAR response at 6 months between smokers and non-smokers. 1.06 (95% CI 0.80–1.41) 
Mattey et al.,111 2009  Prospective  United Kingdom  154  Anti-TNF  At 3 months of treatment a reduction in DAS28 of smokers of 2.07 (29.8%) vs non-smokers 2.64 (38.3%) (P=.008) 
Abhishek et al.,112 2010  RetrospectiveCases-controls  United Kingdom  395  Anti-TNF  At 3 months of treatment good or moderate EULAR response in 17.2% of active smokers vs 36.4% of non-smokers (P=.02) 
Saevarsdottir et al.,109 2011Data from EIRA Cohort  Retrospective  Sweden  535  Anti-TNF  Good EULAR response at 3 months in 29% of active smokers vs 43% non-smokers (P=.03) 
      436  Methotrexate  Good EULAR response at 3 months in 27% of active smokers vs 36% of non-smokers (P=.05) 
Saevarsdottir et al.,121 2011SWEFOT Trial Investigation Group  Retrospective  Sweden  405  Methotrexate  Active smoking predicted non-response according to EULAR at 3–4 months of treatment onset. OR 0.35 (95% CI 0.20–0.63) 

OR: odds ratio; CI: confidence interval.

Tobacco consumption increases the metabolic rate and may be associated with resistance to anti-rheumatic treatment due to pharmacokinetic or pharmacodynamic interactions; for example it has been shown that smokers have lower levels of methotrexate polyglutamates, the active form of the drug, which correlates with clinical117 response. As for the response to anti-TNF differences in its pharmacokinetics or pharmacodynamics due to the use of tobacco have not been reported, although the presence of an increased production of TNF alpha by circulating T cells in smoking patients with RA has been observed, which could justify a lower response to treatment.118

Tobacco and Radiological Progression

The effect of smoking on radiographic progression of RA is not well established. Early studies addressing this aspect.81,8289 were a series of cross-sectional studies in cohorts of long-standing disease (mean 13 years or so) and found a significantly greater radiological progression in patients who smoked. Subsequent publications, both prospective and cross-studies, show conflicting results. In the study by Mattey et al.,119 which analyzes the effect of tobacco in a cohort of 164 patients with RA followed for 5 years, a significantly higher radiological damage Larsen score was seen in smokers compared with nonsmokers (83.1±47.2 and 104.7±49.9, respectively) as was a higher level of disability measured by the HAQ (1.39±0.8 vs 1.77±0.8, respectively). A Greek84 cohort of 287 patients with early onset RA obtained similar results, with greater radiological progression in active and former smokers than in nonsmokers after at least two years of monitoring. However, other prospective studies in large cohorts of recent onset RA do not corroborate these findings and radiological progression of the disease could be influenced by smoking.83,85,87,103,104,120 For example, in a multicenter study in 379 early onset RA patients,120 predictors of radiographic progression were explored in the univariate analysis and the major determinants of radiographic progression were the baseline Larsen score and serological anti-CCP and RF markers and, to a lesser extent, ESR, CRP, age, male gender and being a smoker (OR 1.6, 95% CI 1.0–2.5); however, in the multivariate analysis, smoking was not a independent risk factor, these being the baseline radiological damage, anti-CCP and ESR. Similarly Westhoff et al.85 in their cohort of 894 patients with recent onset RA found a weak association between smoking and radiographic progression, but this was not sustained in multivariate analysis.

However, in most of these studies, patients were not treated evenly, or prognostic factors of radiographic progression markers such as HLA-DRB genotype or the presence of ACPAs not analyzed. Recently our group analyzed, in a cohort of 156 RA patients followed for two years beginning and treated with DMARDs, whether smoking was a predictor of further radiological progression,105 and also analyzed multiple prognostic variables including clinical features, autoantibodies and HLADRB genotype. Multivariate analysis confirmed that active smoking (compared with the absence of that habit) was an independent factor of greater radiological progression assessed by the Larsen index with an OR of 4.3. Other factors for progression were female sex, the presence of HLA-DRB04 and radiographic damage at baseline.

Conclusions

We have studied multiple RA predisposing environmental factors, although there is no doubt that tobacco is established as the most widely studied and with more scientific evidence on his involvement in this disease. Smoking could justify even a third of cases of seropositive RA, as the increased risk associated with tobacco, seems confined primarily to RA with positive autoantibodies (RF and/or ACPAs). The effect of tobacco on the activity of the disease and its clinical course is controversial, as on the progression of joint destruction, yet some studies indicate on a more severe phenotype of RA and greater radiological progression in smokers. Recent studies including large series of patients in registries, such as the Swedish or British, would confirm a worse response to treatment with DMARDs (methotrexate) and/or biological drugs (TNF antagonists) in smokers.

Despite this new evidence on the involvement of smoking in the predisposition and development of RA, there are still issues to resolve, such as the pathogenetic mechanisms involved, the effects of smoking cessation on the disease, the impact its abandonment could have in the general population in the future incidence of RA or its role in other forms of chronic arthritis.121–123 This does not question the fact that clinical rheumatologists must discourage smoking in all patients with RA. New knowledge on tobacco and RA and the fact that smoking is a known risk factor for atherosclerosis and other serious diseases, which are more prevalent in RA patients, fully warrants this advice.

Ethical disclosures

Protection of human and animal subjects. The authors declare that no experiments were performed on humans or animals for this investigation.

Confidentiality of Data. The authors declare that no patient data appears in this article.

Right to privacy and informed consent. The authors declare that no patient data appears in this article.

Conflict of interest

The authors declare no conflict of interest.

References
[1]
M. Heliovaara, K. Aho, A. Aromaa, P. Knekt, A. Reunanen.
Smoking and risk of rheumatoid arthritis.
J Rheumatol, 20 (1993), pp. 1830-1835
[2]
P. Stastny.
Mixed lymphocyte cultures in rheumatoid arthritis.
J Clin Invest, 57 (1976), pp. 1148-1157
[3]
A.J. MacGregor, H. Snieder, A.S. Rigby, M. Koskenvuo, J. Kaprio, K. Aho, et al.
Characterizing the quantitative genetic contribution to rheumatoid arthritis using data from twins.
[4]
M.F. Seldin, C.I. Amos, R. Ward, P.K. Gregersen.
The genetics revolution and the assault on rheumatoid arthritis.
[5]
S. John, A. Myerscough, A. Marlow, A. Hajeer, A. Silman, W. Ollier, et al.
Linkage of cytokine genes to rheumatoid arthritis. Evidence of genetic heterogeneity.
Ann Rheum Dis, 57 (1998), pp. 361-365
[6]
D.L. Mattey, D. Hutchinson.
Smoking and HLA-DR shared epitope alleles in rheumatoid arthritis: comment on the article by Padyukov et al..
Arthritis Rheum, 52 (2005), pp. 3676-3678
[7]
E.W. Karlson, S.C. Chang, J. Cui, L.B. Chibnik, P.A. Fraser, I. De Vivo, et al.
Gene–environment interaction between HLA-DRB1 shared epitope and heavy cigarette smoking in predicting incident rheumatoid arthritis.
Ann Rheum Dis, 69 (2010), pp. 54-60
[8]
H. Kallberg, L. Padyukov, R.M. Plenge, J. Ronnelid, P.K. Gregersen, A.H. van der Helm-van Mil, et al.
Gene–gene and gene–environment interactions involving HLA-DRB1, PTPN22, and smoking in two subsets of rheumatoid arthritis.
Am J Hum Genet, 80 (2007), pp. 867-875
[9]
R.D. Oliveira, C.M. Junta, F.R. Oliveira, L.M. Silva, E.A. Donadi, P. Louzada-Junior.
Share epitope, citrullinated cyclic peptide antibodies and smoking in Brazilian rheumatoid arthritis patients.
Clin Rev Allergy Immunol, 34 (2008), pp. 32-35
[10]
L. Padyukov, C. Silva, P. Stolt, L. Alfredsson, L. Klareskog.
A gene–environment interaction between smoking and shared epitope genes in HLA-DR provides a high risk of seropositive rheumatoid arthritis.
Arthritis Rheum, 50 (2004), pp. 3085-3092
[11]
R. Sanmarti, A. Gomez-Centeno, G. Ercilla, M. Larrosa, O. Vinas, I. Vazquez, et al.
Prognostic factors of radiographic progression in early rheumatoid arthritis: a two year prospective study after a structured therapeutic strategy using DMARDs and very low doses of glucocorticoids.
Clin Rheumatol, 26 (2007), pp. 1111-1118
[12]
F.C. Hall, D.E. Weeks, J.P. Camilleri, L.A. Williams, N. Amos, C. Darke, et al.
Influence of the HLA-DRB1 locus on susceptibility and severity in rheumatoid arthritis.
Q J Med, 89 (1996), pp. 821-829
[13]
C. Turesson, D.J. Schaid, C.M. Weyand, L.T. Jacobsson, J.J. Goronzy, I.F. Petersson, et al.
The impact of HLA-DRB1 genes on extra-articular disease manifestations in rheumatoid arthritis.
Arthritis Res Ther, 7 (2005), pp. R1386-R1393
[14]
F. Cornelis, S. Faure, M. Martinez, J.F. Prud’homme, P. Fritz, C. Dib, et al.
New susceptibility locus for rheumatoid arthritis suggested by a genome-wide linkage study.
Proc Natl Acad Sci USA, 95 (1998), pp. 10746-10750
[15]
D. Jawaheer, P.K. Gregersen.
Rheumatoid arthritis. The genetic components.
Rheum Dis Clin North Am, 28 (2002), pp. 1-15
[16]
A.B. Begovich, V.E. Carlton, L.A. Honigberg, S.J. Schrodi, A.P. Chokkalingam, H.C. Alexander, et al.
A missense single-nucleotide polymorphism in a gene encoding a protein tyrosine phosphatase (PTPN22) is associated with rheumatoid arthritis.
Am J Hum Genet, 75 (2004), pp. 330-337
[17]
A.T. Lee, W. Li, A. Liew, C. Bombardier, M. Weisman, E.M. Massarotti, et al.
The PTPN22 R620W polymorphism associates with RF positive rheumatoid arthritis in a dose-dependent manner but not with HLA-SE status.
Genes Immun, 6 (2005), pp. 129-133
[18]
E.F. Remmers, R.M. Plenge, A.T. Lee, R.R. Graham, G. Hom, T.W. Behrens, et al.
STAT4 and the risk of rheumatoid arthritis and systemic lupus erythematosus.
N Engl J Med, 357 (2007), pp. 977-986
[19]
G. Orozco, B.Z. Alizadeh, A.M. Delgado-Vega, M.A. Gonzalez-Gay, A. Balsa, D. Pascual-Salcedo, et al.
Association of STAT4 with rheumatoid arthritis: a replication study in three European populations.
Arthritis Rheum, 58 (2008), pp. 1974-1980
[20]
R.M. Plenge.
Recent progress in rheumatoid arthritis genetics: one step towards improved patient care.
Curr Opin Rheumatol, 21 (2009), pp. 262-271
[21]
F.A. Kurreeman, L. Padyukov, R.B. Marques, S.J. Schrodi, M. Seddighzadeh, G. Stoeken-Rijsbergen, et al.
A candidate gene approach identifies the TRAF1/C5 region as a risk factor for rheumatoid arthritis.
[22]
J.L. Nelson, K.A. Hughes, A.G. Smith, B.B. Nisperos, A.M. Branchaud, J.A. Hansen.
Maternal–fetal disparity in HLA class II alloantigens and the pregnancy-induced amelioration of rheumatoid arthritis.
N Engl J Med, 329 (1993), pp. 466-471
[23]
C. Jorgensen, M.C. Picot, C. Bologna, J. Sany.
Oral contraception, parity, breast feeding, and severity of rheumatoid arthritis.
Ann Rheum Dis, 55 (1996), pp. 94-98
[24]
E. Berglin, H. Kokkonen, E. Einarsdottir, A. Agren, S. Rantapaa Dahlqvist.
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.
Scand J Rheumatol, 39 (2010), pp. 454-460
[25]
M.F. Doran, C.S. Crowson, W.M. O’Fallon, S.E. Gabriel.
The effect of oral contraceptives and estrogen replacement therapy on the risk of rheumatoid arthritis: a population based study.
J Rheumatol, 31 (2004), pp. 207-213
[26]
P. Brennan, C. Bankhead, A. Silman, D. Symmons.
Oral contraceptives and rheumatoid arthritis: results from a primary care-based incident case-control study.
Semin Arthritis Rheum, 26 (1997), pp. 817-823
[27]
M. Pedersen, S. Jacobsen, M. Klarlund, B.V. Pedersen, A. Wiik, J. Wohlfahrt, et al.
Environmental risk factors differ between rheumatoid arthritis with and without auto-antibodies against cyclic citrullinated peptides.
Arthritis Res Ther, 8 (2006), pp. R133
[28]
E.W. Karlson, L.A. Mandl, S.E. Hankinson, F. Grodstein.
Do breast-feeding and other reproductive factors influence future risk of rheumatoid arthritis? Results from the Nurses’ Health Study.
Arthritis Rheum, 50 (2004), pp. 3458-3467
[29]
M. Pikwer, U. Bergstrom, J.A. Nilsson, L. Jacobsson, G. Berglund, C. Turesson.
Breast feeding, but not use of oral contraceptives, is associated with a reduced risk of rheumatoid arthritis.
Ann Rheum Dis, 68 (2009), pp. 526-530
[30]
M.P. Vessey, L. Villard-Mackintosh, D. Yeates.
Oral contraceptives, cigarette smoking and other factors in relation to arthritis.
Contraception, 35 (1987), pp. 457-464
[31]
M. Pladevall-Vila, G.L. Delclos, C. Varas, H. Guyer, J. Brugues-Tarradellas, A. Anglada-Arisa.
Controversy of oral contraceptives and risk of rheumatoid arthritis: meta-analysis of conflicting studies and review of conflicting meta-analyses with special emphasis on analysis of heterogeneity.
Am J Epidemiol, 144 (1996), pp. 1-14
[32]
M. Pikwer, U. Bergstrom, J.A. Nilsson, L. Jacobsson, C. Turesson.
Early menopause is an independent predictor of rheumatoid arthritis.
Ann Rheum Dis, (2011),
[33]
R. Heikkila, K. Aho, M. Heliovaara, P. Knekt, A. Reunanen, A. Aromaa, et al.
Serum androgen-anabolic hormones and the risk of rheumatoid arthritis.
Ann Rheum Dis, 57 (1998), pp. 281-285
[34]
C. Bengtsson, B. Nordmark, L. Klareskog, I. Lundberg, L. Alfredsson.
Socioeconomic status and the risk of developing rheumatoid arthritis: results from the Swedish EIRA study.
Ann Rheum Dis, 64 (2005), pp. 1588-1594
[35]
M. Pedersen, S. Jacobsen, M. Klarlund, M. Frisch.
Socioeconomic status and risk of rheumatoid arthritis: a Danish case-control study.
J Rheumatol, 33 (2006), pp. 1069-1074
[36]
U. Bergstrom, L.T. Jacobsson, J.A. Nilsson, G. Berglund, C. Turesson.
Pulmonary dysfunction, smoking, socioeconomic status and the risk of developing rheumatoid arthritis.
Rheumatology (Oxford), 50 (2011), pp. 2005-2013
[37]
M. Rosell, A.M. Wesley, K. Rydin, L. Klareskog, L. Alfredsson.
Dietary fish and fish oil and the risk of rheumatoid arthritis.
Epidemiology, 20 (2009), pp. 896-901
[38]
D.J. Pattison, R.A. Harrison, D.P. Symmons.
The role of diet in susceptibility to rheumatoid arthritis: a systematic review.
J Rheumatol, 31 (2004), pp. 1310-1319
[39]
E. Benito-Garcia, D. Feskanich, F.B. Hu, L.A. Mandl, E.W. Karlson.
Protein, iron, and meat consumption and risk for rheumatoid arthritis: a prospective cohort study.
Arthritis Res Ther, 9 (2007), pp. R16
[40]
M.M. Nielen, D. van Schaardenburg, W.F. Lems, R.J. van de Stadt, M.H. de Koning, H.W. Reesink, et al.
Vitamin D deficiency does not increase the risk of rheumatoid arthritis: comment on the article by Merlino et al..
Arthritis Rheum, 54 (2006), pp. 3719-3720
[41]
M.F. Holick.
Vitamin D deficiency.
N Engl J Med, 357 (2007), pp. 266-281
[42]
L.A. Merlino, J. Curtis, T.R. Mikuls, J.R. Cerhan, L.A. Criswell, K.G. Saag.
Vitamin D intake is inversely associated with rheumatoid arthritis: results from the Iowa Women's Health Study.
Arthritis Rheum, 50 (2004), pp. 72-77
[43]
S. Patel, T. Farragher, J. Berry, D. Bunn, A. Silman, D. Symmons.
Association between serum vitamin D metabolite levels and disease activity in patients with early inflammatory polyarthritis.
Arthritis Rheum, 56 (2007), pp. 2143-2149
[44]
U.J. Haque, S.J. Bartlett.
Relationships among vitamin D, disease activity, pain and disability in rheumatoid arthritis.
Clin Exp Rheumatol, 28 (2010), pp. 745-747
[45]
G.S. Kerr, I. Sabahi, J.S. Richards, L. Caplan, G.W. Cannon, A. Reimold, et al.
Prevalence of vitamin D insufficiency/deficiency in rheumatoid arthritis and associations with disease severity and activity.
J Rheumatol, 38 (2011), pp. 53-59
[46]
H. Kallberg, S. Jacobsen, C. Bengtsson, M. Pedersen, L. Padyukov, P. Garred, et al.
Alcohol consumption is associated with decreased risk of rheumatoid arthritis: results from two Scandinavian case-control studies.
Ann Rheum Dis, 68 (2009), pp. 222-227
[47]
E.W. Karlson, L.A. Mandl, G.N. Aweh, F. Grodstein.
Coffee consumption and risk of rheumatoid arthritis.
Arthritis Rheum, 48 (2003), pp. 3055-3060
[48]
T.R. Mikuls, J.R. Cerhan, L.A. Criswell, L. Merlino, A.S. Mudano, M. Burma, et al.
Coffee, tea, and caffeine consumption and risk of rheumatoid arthritis: results from the Iowa Women's Health Study.
[49]
M. Heliovaara, K. Aho, P. Knekt, O. Impivaara, A. Reunanen, A. Aromaa.
Coffee consumption, rheumatoid factor, and the risk of rheumatoid arthritis.
Ann Rheum Dis, 59 (2000), pp. 631-635
[50]
P. de Pablo, T. Dietrich, T.E. McAlindon.
Association of periodontal disease and tooth loss with rheumatoid arthritis in the US population.
J Rheumatol, 35 (2008), pp. 70-76
[51]
K. Lundberg, N. Wegner, T. Yucel-Lindberg, P.J. Venables.
Periodontitis in RA – the citrullinated enolase connection.
Nat Rev Rheumatol, 6 (2010), pp. 727-730
[52]
E.D. Rosenstein, R.A. Greenwald, L.J. Kushner, G. Weissmann.
Hypothesis: the humoral immune response to oral bacteria provides a stimulus for the development of rheumatoid arthritis.
Inflammation, 28 (2004), pp. 311-318
[53]
H. Marotte, P. Farge, P. Gaudin, C. Alexandre, B. Mougin, P. Miossec.
The association between periodontal disease and joint destruction in rheumatoid arthritis extends the link between the HLA-DR shared epitope and severity of bone destruction.
Ann Rheum Dis, 65 (2006), pp. 905-909
[54]
P. Stolt, A. Yahya, C. Bengtsson, H. Kallberg, J. Ronnelid, I. Lundberg, et al.
Silica exposure among male current smokers is associated with a high risk of developing ACPA-positive rheumatoid arthritis.
Ann Rheum Dis, 69 (2010), pp. 1072-1076
[55]
J.M. Hazes, B.A. Dijkmans, J.P. Vandenbroucke, R.R. de Vries, A. Cats.
Lifestyle and the risk of rheumatoid arthritis: cigarette smoking and alcohol consumption.
Ann Rheum Dis, 49 (1990), pp. 980-982
[56]
M. Hernandez Avila, M.H. Liang, W.C. Willett, M.J. Stampfer, G.A. Colditz, B. Rosner, et al.
Reproductive factors, smoking, and the risk for rheumatoid arthritis.
Epidemiology, 1 (1990), pp. 285-291
[57]
D.P. Symmons, C.R. Bankhead, B.J. Harrison, P. Brennan, E.M. Barrett, D.G. Scott, 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.
[58]
E.W. Karlson, I.M. Lee, N.R. Cook, J.E. Manson, J.E. Buring, C.H. Hennekens.
A retrospective cohort study of cigarette smoking and risk of rheumatoid arthritis in female health professionals.
[59]
M.L. Sopori, W. Kozak.
Immunomodulatory effects of cigarette smoke.
J Neuroimmunol, 83 (1998), pp. 148-156
[60]
Y. Arnson, Y. Shoenfeld, H. Amital.
Effects of tobacco smoke on immunity, inflammation and autoimmunity.
J Autoimmun, 34 (2010), pp. J258-J265
[61]
J. Majo, H. Ghezzo, M.G. Cosio.
Lymphocyte population and apoptosis in the lungs of smokers and their relation to emphysema.
Eur Respir J, 17 (2001), pp. 946-953
[62]
W.A. Pryor, K. Stone, L.Y. Zang, E. Bermudez.
Fractionation of aqueous cigarette tar extracts: fractions that contain the tar radical cause DNA damage.
Chem Res Toxicol, 11 (1998), pp. 441-448
[63]
M.K. Formica, J.R. Palmer, L. Rosenberg, T.E. McAlindon.
Smoking, alcohol consumption, and risk of systemic lupus erythematosus in the Black Women's Health Study.
J Rheumatol, 30 (2003), pp. 1222-1226
[64]
A.A. Bengtsson, L. Rylander, L. Hagmar, O. Nived, G. Sturfelt.
Risk factors for developing systemic lupus erythematosus: a case-control study in southern Sweden.
Rheumatology (Oxford), 41 (2002), pp. 563-571
[65]
M.A. Hernan, M.J. Olek, A. Ascherio.
Cigarette smoking and incidence of multiple sclerosis.
Am J Epidemiol, 154 (2001), pp. 69-74
[66]
T. Riise, M.W. Nortvedt, A. Ascherio.
Smoking is a risk factor for multiple sclerosis.
Neurology, 61 (2003), pp. 1122-1124
[67]
I.A. Holm, J.E. Manson, K.B. Michels, E.K. Alexander, W.C. Willett, R.D. Utiger.
Smoking and other lifestyle factors and the risk of Graves’ hyperthyroidism.
Arch Intern Med, 165 (2005), pp. 1606-1611
[68]
P. Vestergaard, L. Rejnmark, J. Weeke, H.C. Hoeck, H.K. Nielsen, J. Rungby, et al.
Smoking as a risk factor for Graves’ disease, toxic nodular goiter, and autoimmune hypothyroidism.
[69]
M.E. Gershwin, C. Selmi, H.J. Worman, E.B. Gold, M. Watnik, J. Utts, et al.
Risk factors and comorbidities in primary biliary cirrhosis: a controlled interview-based study of 1032 patients.
Hepatology, 42 (2005), pp. 1194-1202
[70]
D. Hutchinson, L. Shepstone, R. Moots, J.T. Lear, M.P. Lynch.
Heavy cigarette smoking is strongly associated with rheumatoid arthritis (RA), particularly in patients without a family history of RA.
Ann Rheum Dis, 60 (2001), pp. 223-227
[71]
E. Krishnan.
Smoking, gender and rheumatoid arthritis – epidemiological clues to etiology. Results from the behavioral risk factor surveillance system.
Joint Bone Spine, 70 (2003), pp. 496-502
[72]
E. Krishnan, T. Sokka, P. Hannonen.
Smoking–gender interaction and risk for rheumatoid arthritis.
Arthritis Res Ther, 5 (2003), pp. R158-R162
[73]
L.A. Criswell, L.A. Merlino, J.R. Cerhan, T.R. Mikuls, A.S. Mudano, M. Burma, et al.
Cigarette smoking and the risk of rheumatoid arthritis among postmenopausal women: results from the Iowa Women's Health Study.
Am J Med, 112 (2002), pp. 465-471
[74]
K.H. Costenbader, D. Feskanich, L.A. Mandl, E.W. Karlson.
Smoking intensity, duration, and cessation, and the risk of rheumatoid arthritis in women.
[75]
P. Stolt, C. Bengtsson, B. Nordmark, S. Lindblad, I. Lundberg, L. Klareskog, et al.
Quantification of the influence of cigarette smoking on rheumatoid arthritis: results from a population based case-control study, using incident cases.
Ann Rheum Dis, 62 (2003), pp. 835-841
[76]
L.F. Voigt, T.D. Koepsell, J.L. Nelson, C.E. Dugowson, J.R. Daling.
Smoking, obesity, alcohol consumption, and the risk of rheumatoid arthritis.
Epidemiology, 5 (1994), pp. 525-532
[77]
A. Reckner Olsson, T. Skogh, G. Wingren.
Comorbidity and lifestyle, reproductive factors, and environmental exposures associated with rheumatoid arthritis.
Ann Rheum Dis, 60 (2001), pp. 934-939
[78]
A.R. Olsson, T. Skogh, G. Wingren.
Aetiological factors of importance for the development of rheumatoid arthritis.
Scand J Rheumatol, 33 (2004), pp. 300-306
[79]
D. Sugiyama, K. Nishimura, K. Tamaki, G. Tsuji, T. Nakazawa, A. Morinobu, et al.
Impact of smoking as a risk factor for developing rheumatoid arthritis: a meta-analysis of observational studies.
Ann Rheum Dis, 69 (2010), pp. 70-81
[80]
H. Kallberg, B. Ding, L. Padyukov, C. Bengtsson, J. Ronnelid, L. Klareskog, et al.
Smoking is a major preventable risk factor for rheumatoid arthritis: estimations of risks after various exposures to cigarette smoke.
Ann Rheum Dis, 70 (2011), pp. 508-511
[81]
K.G. Saag, J.R. Cerhan, S. Kolluri, K. Ohashi, G.W. Hunninghake, D.A. Schwartz.
Cigarette smoking and rheumatoid arthritis severity.
Ann Rheum Dis, 56 (1997), pp. 463-469
[82]
F. Wolfe.
The effect of smoking on clinical, laboratory, and radiographic status in rheumatoid arthritis.
J Rheumatol, 27 (2000), pp. 630-637
[83]
V.F. Manfredsdottir, T. Vikingsdottir, T. Jonsson, A.J. Geirsson, O. Kjartansson, M. Heimisdottir, et al.
The effects of tobacco smoking and rheumatoid factor seropositivity on disease activity and joint damage in early rheumatoid arthritis.
Rheumatology (Oxford), 45 (2006), pp. 734-740
[84]
N.G. Papadopoulos, Y. Alamanos, P.V. Voulgari, E.K. Epagelis, N. Tsifetaki, A.A. Drosos.
Does cigarette smoking influence disease expression, activity and severity in early rheumatoid arthritis patients?.
Clin Exp Rheumatol, 23 (2005), pp. 861-866
[85]
G. Westhoff, R. Rau, A. Zink.
Rheumatoid arthritis patients who smoke have a higher need for DMARDs and feel worse, but they do not have more joint damage than non-smokers of the same serological group.
Rheumatology (Oxford), 47 (2008), pp. 849-854
[86]
M. Soderlin, I. Petersson, S. Bergman, B. Svensson.
Smoking at onset of rheumatoid arthritis (RA) and its effect on disease activity and functional status: experiences from BARFOT, a long-term observational study on early RA.
Scand J Rheumatol, 40 (2011), pp. 249-255
[87]
T.R. Mikuls, L.B. Hughes, A.O. Westfall, V.M. Holers, L. Parrish, D. van der Heijde, et al.
Cigarette smoking, disease severity and autoantibody expression in African Americans with recent-onset rheumatoid arthritis.
Ann Rheum Dis, 67 (2008), pp. 1529-1534
[88]
H.S. Lee, P. Irigoyen, M. Kern, A. Lee, F. Batliwalla, H. Khalili, 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 Rheum, 56 (2007), pp. 1745-1753
[89]
B. Masdottir, T. Jonsson, V. Manfredsdottir, A. Vikingsson, A. Brekkan, H. Valdimarsson.
Smoking, rheumatoid factor isotypes and severity of rheumatoid arthritis.
Rheumatology (Oxford), 39 (2000), pp. 1202-1205
[90]
L. Klareskog, P. Stolt, K. Lundberg, H. Kallberg, C. Bengtsson, J. Grunewald, 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 Rheum, 54 (2006), pp. 38-46
[91]
D.M. Lee, R. Phillips, E.M. Hagan, L.B. Chibnik, K.H. Costenbader, P.H. Schur.
Quantifying anti-cyclic citrullinated peptide titres: clinical utility and association with tobacco exposure in patients with rheumatoid arthritis.
Ann Rheum Dis, 68 (2009), pp. 201-208
[92]
L. Michou, V.H. Teixeira, C. Pierlot, S. Lasbleiz, T. Bardin, P. Dieude, et al.
Associations between genetic factors, tobacco smoking and autoantibodies in familial and sporadic rheumatoid arthritis.
Ann Rheum Dis, 67 (2008), pp. 466-470
[93]
S.P. Linn-Rasker, A.H. van der Helm-van Mil, F.A. van Gaalen, M. Kloppenburg, R.R. de Vries, S. le Cessie, et al.
Smoking is a risk factor for anti-CCP antibodies only in rheumatoid arthritis patients who carry HLA-DRB1 shared epitope alleles.
Ann Rheum Dis, 65 (2006), pp. 366-371
[94]
M. Pedersen, S. Jacobsen, P. Garred, H.O. Madsen, M. Klarlund, A. Svejgaard, et al.
Strong combined gene–environment effects in anti-cyclic citrullinated peptide-positive rheumatoid arthritis: a nationwide case-control study in Denmark.
Arthritis Rheum, 56 (2007), pp. 1446-1453
[95]
A.H. van der Helm-van Mil, K.N. Verpoort, S. le Cessie, T.W. Huizinga, R.R. de Vries, R.E. Toes.
The HLA-DRB1 shared epitope alleles differ in the interaction with smoking and predisposition to antibodies to cyclic citrullinated peptide.
Arthritis Rheum, 56 (2007), pp. 425-432
[96]
D. Makrygiannakis, M. Hermansson, A.K. Ulfgren, A.P. Nicholas, A.J. Zendman, A. Eklund, et al.
Smoking increases peptidylarginine deiminase 2 enzyme expression in human lungs and increases citrullination in BAL cells.
Ann Rheum Dis, 67 (2008), pp. 1488-1492
[97]
D. Baeten, I. Peene, A. Union, L. Meheus, M. Sebbag, G. Serre, et al.
Specific presence of intracellular citrullinated proteins in rheumatoid arthritis synovium: relevance to antifilaggrin autoantibodies.
Arthritis Rheum, 44 (2001), pp. 2255-2262
[98]
L. Klareskog, J. Ronnelid, K. Lundberg, L. Padyukov, L. Alfredsson.
Immunity to citrullinated proteins in rheumatoid arthritis.
Annu Rev Immunol, 26 (2008), pp. 651-675
[99]
S. Rantapaa-Dahlqvist, B.A. de Jong, E. Berglin, G. Hallmans, G. Wadell, H. Stenlund, et al.
Antibodies against cyclic citrullinated peptide and IgA rheumatoid factor predict the development of rheumatoid arthritis.
Arthritis Rheum, 48 (2003), pp. 2741-2749
[100]
M.M. Nielen, D. van Schaardenburg, H.W. Reesink, R.J. van de Stadt, I.E. van der Horst-Bruinsma, M.H. de Koning, et al.
Specific autoantibodies precede the symptoms of rheumatoid arthritis: a study of serial measurements in blood donors.
Arthritis Rheum, 50 (2004), pp. 380-386
[101]
V. Ruiz-Esquide, M.J. Gómara, V.I. Peinado, J.A. Gómez-Puerta, J.A. Barberá, J.D. Cãnete, et al.
Anti-citrullinated peptide antibodies in the serum of heavy smokers without rheumatoid arthritis. A differential effect of chronic obstructive pulmonary disease?.
[102]
A.M. Wood, P. de Pablo, C.D. Buckley, A. Ahmad, R.A. Stockley.
Smoke exposure as a determinant of autoantibody titre in alpha-antitrypsin deficiency and COPD.
Eur Respir J, 37 (2011), pp. 32-38
[103]
B.J. Harrison, A.J. Silman, N.J. Wiles, D.G. Scott, D.P. Symmons.
The association of cigarette smoking with disease outcome in patients with early inflammatory polyarthritis.
[104]
A. Finckh, S. Dehler, K.H. Costenbader, C. Gabay.
Cigarette smoking and radiographic progression in rheumatoid arthritis.
Ann Rheum Dis, 66 (2007), pp. 1066-1071
[105]
V. Ruiz-Esquide, J.A. Gomez-Puerta, J.D. Canete, E. Graell, I. Vazquez, M.G. Ercilla, et al.
Effects of smoking on disease activity and radiographic progression in early rheumatoid arthritis.
J Rheumatol, 38 (2011), pp. 2536-2539
[106]
C. Salliot, K. Dawidowicz, C. Lukas, M. Guedj, C. Paccard, J. Benessiano, et al.
PTPN22 R620W genotype–phenotype correlation analysis and gene–environment interaction study in early rheumatoid arthritis: results from the ESPOIR cohort.
Rheumatology (Oxford), 50 (2011), pp. 1802-1808
[107]
J. Rojas-Serrano, L.L. Perez, C.G. Garcia, F. Moctezuma, E. Alvarez-Hernandez, J. Vazquez-Mellado, et al.
Current smoking status is associated to a non-ACR 50 response in early rheumatoid arthritis. A cohort study.
Clin Rheumatol, 30 (2011), pp. 1589-1593
[108]
C. Turesson, W.M. O’Fallon, C.S. Crowson, S.E. Gabriel, E.L. Matteson.
Extra-articular disease manifestations in rheumatoid arthritis: incidence trends and risk factors over 46 years.
Ann Rheum Dis, 62 (2003), pp. 722-727
[109]
K.G. Saag, S. Kolluri, R.K. Koehnke, T.A. Georgou, J.W. Rachow, G.W. Hunninghake, et al.
Rheumatoid arthritis lung disease. Determinants of radiographic and physiologic abnormalities.
Arthritis Rheum, 39 (1996), pp. 1711-1719
[110]
H.K. Lee, D.S. Kim, B. Yoo, J.B. Seo, J.Y. Rho, T.V. Colby, et al.
Histopathologic pattern and clinical features of rheumatoid arthritis-associated interstitial lung disease.
Chest, 127 (2005), pp. 2019-2027
[111]
J.A. Wessels, S.M. van der Kooij, S. le Cessie, W. Kievit, P. Barerra, C.F. Allaart, et al.
A clinical pharmacogenetic model to predict the efficacy of methotrexate monotherapy in recent-onset rheumatoid arthritis.
Arthritis Rheum, 56 (2007), pp. 1765-1775
[112]
S. Saevarsdottir, S. Wedren, M. Seddighzadeh, C. Bengtsson, A. Wesley, S. Lindblad, 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 Rheum, 63 (2011), pp. 26-36
[113]
K.L. Hyrich, K.D. Watson, A.J. Silman, D.P. Symmons.
Predictors of response to anti-TNF-alpha therapy among patients with rheumatoid arthritis: results from the British Society for Rheumatology Biologics Register.
Rheumatology (Oxford), 45 (2006), pp. 1558-1565
[114]
D.L. Mattey, A. Brownfield, P.T. Dawes.
Relationship between pack-year history of smoking and response to tumor necrosis factor antagonists in patients with rheumatoid arthritis.
J Rheumatol, 36 (2009), pp. 1180-1187
[115]
A. Abhishek, S. Butt, K. Gadsby, W. Zhang, C.M. Deighton.
Anti-TNF-alpha agents are less effective for the treatment of rheumatoid arthritis in current smokers.
J Clin Rheumatol, 16 (2010), pp. 15-18
[116]
M. van den Broek, N.B. Klarenbeek, L. Dirven, D. van Schaardenburg, H.M. Hulsmans, P.J. Kerstens, et al.
Discontinuation of infliximab and potential predictors of persistent low disease activity in patients with early rheumatoid arthritis and disease activity score-steered therapy: subanalysis of the BeSt study.
Ann Rheum Dis, 70 (2011), pp. 1389-1394
[117]
L.K. Stamp, J.L. O’Donnell, P.T. Chapman, M. Zhang, C. Frampton, J. James, et al.
Determinants of red blood cell methotrexate polyglutamate concentrations in rheumatoid arthritis patients receiving long-term methotrexate treatment.
Arthritis Rheum, 60 (2009), pp. 2248-2256
[118]
J.R. Glossop, P.T. Dawes, D.L. Mattey.
Association between cigarette smoking and release of tumour necrosis factor alpha and its soluble receptors by peripheral blood mononuclear cells in patients with rheumatoid arthritis.
Rheumatology (Oxford), 45 (2006), pp. 1223-1229
[119]
D.L. Mattey, D. Hutchinson, P.T. Dawes, N.B. Nixon, S. Clarke, J. Fisher, et al.
Smoking and disease severity in rheumatoid arthritis: association with polymorphism at the glutathione S-transferase M1 locus.
Arthritis Rheum, 46 (2002), pp. 640-646
[120]
K. Forslind, M. Ahlmen, K. Eberhardt, I. Hafstrom, B. Svensson.
Prediction of radiological outcome in early rheumatoid arthritis in clinical practice: role of antibodies to citrullinated peptides (anti-CCP).
Ann Rheum Dis, 63 (2004), pp. 1090-1095
[121]
H.Y. Chung, P. Machado, D. van der Heijde, M.A. D’Agostino, M. Dougados.
Smokers in early axial spondyloarthritis have earlier disease onset, more disease activity, inflammation and damage, and poorer function and health-related quality of life: results from the DESIR cohort.
Ann Rheum Dis, (2011),
[122]
D. Poddubnyy, H. Haibel, J. Listing, E. Marker-Hermann, H. Zeidler, J. Braun, et al.
Baseline radiographic damage, elevated acute phase reactants and cigarette smoking status predict radiographic progression in the spine in early axial spondyloarthritis.
Arthritis Rheum, (2011),
[123]
L. Eder, T. Law, V. Chandran, S. Shanmugarajah, H. Shen, C.F. Rosen, et al.
Association between environmental factors and onset of psoriatic arthritis in patients with psoriasis.
Arthritis Care Res (Hoboken), 63 (2011), pp. 1091-1097

Please cite this article as: Ruiz-Esquide V, Sanmartí R. Tabaco y otros factores ambientales en la artritis reumatoide. Reumatol Clin. 2012;8:342–50.

Copyright © 2011. Elsevier España, S.L.. All rights reserved
Idiomas
Reumatología Clínica (English Edition)
Article options
Tools
es en

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?