Smoking associates with increased BAFF and decreased interferon-γ levels in patients with systemic lupus erythematosus ======================================================================================================================= * Warren David Raymond * Matthew Hamdorf * Michael Furfaro * Gro Ostli Eilertsen * Johannes Cornelis Nossent ## Abstract **Objective** In SLE, smoking increases the burden of cutaneous disease and organ damage, and leads to premature mortality. However, the effect of smoking on disease manifestations and cytokine levels of patients with SLE is unclear. This study compared characteristics of patients with SLE across smoking status, and determined the association of smoking with serum cytokine levels. **Method** A cross-sectional study of patients with SLE (n=99) during a research visit in which smoking status was ascertained. Smoking status was compared across classification criteria (American College of Rheumatology Classification Criteria for SLE (ACR97)), disease activity (SLE Disease Activity Index), autoantibody levels, accrued damage (Systemic Lupus International Collaborating Clinics/ACR Damage Index), and circulating concentrations of serum interferon-gamma (IFN-γ), interleukin (IL)-1β, IL-4, IL-6, IL-10, IL-12, IL-17, B cell-activating factor (BAFF), tumour necrosis factor-alpha, transforming growth factor beta 1 (TGF-β1), macrophage inflammatory protein 1 alpha (MIP-1α), MIP-1β and monocyte chemoattractant protein 1. Linear regression models determined the association between smoking and cytokine levels, adjusting for age and sex, clinical characteristics (model 1), and anti-inflammatory (IL-4, IL-10 and TGF- β1) and regulatory (IL-1β) cytokines (model 2). **Results** Among patients with SLE (97.9% ANA+; mean 48.48 years old; 86.9% female; mean 10 years of disease duration), 35.4% (n=35 of 99) were smoking (an average of 7 cigarettes/day for 24 years). Smokers had increased odds of prevalent ACR97 malar rash (OR 3.40, 95% CI 1.23 to 9.34) and mucosal ulcers (OR 3.31, 95% CI 1.36 to 8.05). Smokers had more arthritis (OR 3.19, 95% CI 1.19 to 8.60), migraine (OR 2.82, 95% CI 1.07 to 7.44), Raynaud’s phenomenon (OR 5.15, 95% CI 1.95 to 13.56) and increased non-steroidal anti-inflammatory drug use (OR 6.88, 95% CI 1.99 to 23.72). Smoking associated with 27% increased BAFF levels (95% CI 6% to 48%) and 42% decreased IFN-γ levels (95% CI −79% to −5%) in model 2. **Conclusion** In patients with SLE, smoking independently associated with increased BAFF and decreased IFN-γ levels, and an increased frequency of arthritis, migraine and Raynaud’s phenomenon. Smoking cessation is advisable to reduce systemic inflammation, reduce disease activity and improve host defence. * cytokines * lupus erythematosus * systemic * smoking ### Key messages #### What is already known about this subject? * Patients with SLE who smoke have more cutaneous disease, organ damage and premature mortality, as well as reduced medication efficacy. However, the effects of smoking on SLE disease activity and circulating cytokine levels are unclear. #### What does this study add? * This study shows that patients with SLE who smoke have increased odds of arthritis, migraine (lupus headache) and secondary Raynaud’s phenomenon, which necessitated additional non-steroidal anti-inflammatory drug use. * This study showed that while smoking had a broadly immunosuppressive effect on cytokine levels in patients with SLE, it was independently associated with increased B cell-activating levels and decreased interferon-gamma (IFN-γ) levels. #### How might this impact on clinical practice or future developments? * Smoking cessation should be advised to reduce systemic inflammation (B cell-activating factor levels) and disease activity, that is, arthritis, vasospasm (Raynaud’s phenomenon), vasoconstriction (migraine/headache), and global disease activity scores [patient/physician visual analogue scale(VAS)], as well as improve host defence (IFN-γ) in patients with SLE. ## Introduction SLE is the prototypical autoimmune disease characterised by chronic, multisystem inflammation.1 Patients experience an unpredictable disease course, which leads to end-stage organ damage and premature mortality.1 2 Epidemiological evidence suggests that smoking contributes to the development of ANA and dsDNA autoantibody formation, which are central to the pathogenesis of SLE, particularly lupus nephritis (LN)3; and smoking has been linked to an increased risk of developing a range of autoimmune diseases, including SLE.4–6 In patients with SLE, smoking is linked to cutaneous manifestations,7 damage accrual, including the earlier onset of end-stage renal disease in those with LN,8 and premature mortality.9 10 However, the association between smoking and SLE disease activity is relatively understudied.11 Cigarette smoke exposes the epithelial cells of the larynx, bronchi and lung to more than 60 chemical carcinogens, which each has the potential to cause DNA damage.12 Furthermore, smoking has been shown to increase and decrease many proinflammatory and anti-inflammatory cytokines in the general population, with13 and without SARS-CoV-2 (COVID-19),14 and in a cohort with Sjogren’s syndrome.15 Yet, there are (very) limited data on the impact of smoking on serum cytokines in patients with SLE, especially in the context of medication use and organ damage.5 In SLE, cytokines such as B-cell activating factor (BAFF),16 transforming growth factor beta 1 (TGF-β1)17 and interferons (IFNs)18 are associated with disease activity and severity. Therefore, if smoking were to exacerbate abnormal levels of these cytokines, it would have implications on the treatment and management of SLE. Thus, in this study, we aimed to describe the impact of smoking with a range of clinical, serological and immunological characteristics in a cohort of patients with SLE; and to determine the association between smoking and cytokine levels adjusting for age, sex, medication use, disease activity and organ damage. ## Method This is a cross-sectional study of 99 patients with SLE fulfilling the American College of Rheumatology’s (ACR) classification criteria for SLE.19 20 Smoking was defined as the current consumption of either prefabricated or hand-rolled cigarettes at the research visit. Ex-smokers (median 10 years since quitting and a median 3.5 pack-year exposure) were counted as non-smoking. This was done to align with the stronger association of current smoking rather than ex-smoking or non-smoking behaviours with disease activity and severity in patients with SLE.21 22 Clinical data including disease activity and laboratory results were collected at a research visit, where serums taken were stored within 2 hours at −70°C. Cytokine assays were performed for all participants at the same time. Serological, immunological and biochemistry levels were measured in an accredited laboratory on samples taken at the time of research visit. Disease activity was recorded using the SLE Disease Activity Index-2K (SLEDAI-2K).23 We calculated Lupus Low Disease Activity State (LLDAS) scores over time,24 and presented these data as either >30%, >50%, >70% of their time spent in an LLDAS as per Sharma *et al*.25 Damage accrual was captured prospectively for each participant with the Systemic Lupus International Collaborating Clinics/ACR Damage Index for SLE (SDI) during a median follow-up of 10 years from date of SLE diagnosis through to the research visit.26 Medication data were collected at the research visit, which documented the use of prednisone (oral or intravenous methylprednisone), cytostatic agents (azathioprine, cyclophosphamide or mycophenolate) or immunomodulators (hydroxychloroquine or rituximab). Cytokines (IFN-gamma (IFN-γ), interleukin (IL)-1β (IL-1β), IL-4, IL-6, IL-10, IL-12, IL-17A, BAFF, macrophage inflammatory protein 1 alpha (MIP-1α) (patients with SLE only), MIP 1 beta (MIP-1β) (patients with SLE only), monocyte chemoattractant protein 1 (MCP-1), tumour necrosis factor-alpha (TNF-α) and TGF-β1) were measured in stored (−70°C) serum samples by a quantitative sandwich immunoassay (Single Analyte ELISArray kit; SuperArray Bioscience Corp, Frederick, Maryland, USA). All assays were run in duplicate and the results averaged. The manufacturer’s recommendations were followed throughout, and the same lot was used for each cytokine. For statistical purposes, values below the limit of detection (LOD) were replaced by the LOD value. ### Statistical methods Continuous clinical, serological, and immunological variables are described as a mean±SD or median and IQR depending on the data distribution. Continuous data were compared across smoking status with t-test, Mann-Whitney U test, one-way analysis of variance (ANOVA) or Kruskal-Wallis test. Categorical clinical, serological, and immunological variables are described as frequency (n) and proportion (%), and compared across smoking status with logistic regression (presented as an OR with 95% CIs or a Fisher’s exact test). Cytokine levels were described with a geometric mean and 95% CI after undergoing log-transformed to improve normality and then back-transformed with Euler’s number. The associations between smoking and proinflammatory cytokines were determined using Spearman correlation coefficients (Rs) and age-adjusted and sex-adjusted linear regression models. Additional multiple regression modelling of the association between smoking and cytokine levels included clinical characteristics (age, sex, prednisone use, immunosuppressant use, SLEDAI score and SDI score) which are known to influence cytokine levels (model 1). Finally, we performed an ad-hoc multiple regression model which determined the association of smoking with serum BAFF, IFN-γ, IL-12, IL-17, TNF-α or MIP-1β, adjusted for model 1 plus anti-inflammatory cytokines (IL-4, IL-10, TGF-β1) and the regulatory cytokine (IL-1β). This was to determine whether the minimally adjusted association(s) between smoking and cytokine levels held after accounting for known lower TGF-β1 levels in patients with SLE,17 and to account for the immunosuppressive effects of smoking on key regulatory and anti-inflammatory cytokines IL-1β, IL-4 and IL-10.27–30 ## Results ### Clinical characteristics of patients with SLE at the research visit compared across smoking status At the research visit, patients with SLE (97.9% ANA+) were on average 48 years old, 86.9% female and had been followed up for an average of 10 years. At the research visit, 35.4% (n=35 of 99) were currently smoking (average of 7 cigarettes per day for 24 years) (table 1). Within non-smokers, 39.1% (n=25 of 64) were ex-smokers (average of 9 cigarettes per day for 15 years), with cessation occurring on average 10 years prior to the research visit. Current smokers had a significantly higher pack-year smoking exposure than ex-smokers (9 vs 3.5 pack-years, p=0.003). Within current smokers, 57.1% (n=20 of 35) had a <10 pack-year smoking exposure. A total of 34.3% (n=12 of 35) had a 10–20 pack-year smoking exposure, and 8.6% (n=3 of 35) had a ≥20 pack-year smoking exposure. Current and non-smokers had an equivalent burden of comorbid vascular, pulmonary, malignancy and metabolic pathology, as well as accrued damage (SDI >0) and total SDI scores. However, patients with any smoking exposure (current and/or ex-smokers) had higher accrued damage (69.0% vs 46.3%; OR 2.57, 95% CI 1.12 to 5.89; p=0.025) and malignancy (20.7% vs 4.9%; OR 5.09, 95% CI 1.07 to 24.13; p=0.041). View this table: [Table 1](http://lupus.bmj.com/content/8/1/e000537/T1) Table 1 Characteristics of patients with SLE at the research visit compared across smoking status There was a higher cumulative prevalence of malar rash (OR 3.40, 95% CI 1.23 to 9.34; p=0.018), mucosal ulcers (OR 3.31, 95% CI 1.36 to 8.05; p=0.008), arthritis (OR 3.19, 95% CI 1.19 to 8.60; p=0.022) and Raynaud’s phenomenon (OR 5.15, 95% CI 1.95 to 13.56; p=0.001) in current smokers (online supplemental table 1). SLEDAI-2K scores were equivalent across smoking status, although current smokers had increased odds of migraine (OR 2.82, 95% CI 1.07 to 7.44; p=0.037), with similar representation of other SLEDAI features. Both patient (4 vs 2, p=0.003) and physician (3 vs 2, p=0.046) global disease activity (GDA) visual analogue scale (VAS) scores were however higher. The increased odds of Raynaud’s phenomenon in current smokers were higher in male patients (data not shown). Laboratory findings demonstrated that current and non-smokers had similar immunological disease activity, including ANA and anti-dsDNA seropositivity, hypocomplementemia and cytopenias. However, current smokers had higher levels of haemoglobin (13.06 vs 12.99, p=0.036), mean corpuscular volume (MCV) (91.91 vs 88.52, p=0.014) and B cells (0.14 vs 0.08, p=0.037), with lower ApoA 1 lipoprotein levels (1.48 vs 1.61, p=0.039). Smoking was not associated with anti-dsDNA titres (ELIA Rs −0.03, p=0.776), complement protein (C3) (Rs −0.07, p=0.480) or C4 (Rs 0.14, p=0.174) (online supplemental table 4). ### Supplementary data [[lupus-2021-000537supp001.pdf]](pending:yes) The management of SLE required non-steroidal anti-inflammatories in 15.2% (n=15 of 99), prednisone in 50.5% (50 of 99), hydroxychloroquine in 58.6% (n=58 of 99) and immunosuppression in 36.4% (n=36 of 99). Medication requirement for SLE was similar across smoking status with the exception of additional non-steroidal anti-inflammatory drug (NSAID) requirement in smokers (OR 6.88, 95% CI 1.99 to 23.72; p=0.002). Medication requirement for comorbid conditions was equivalent across smoking status (online supplemental table 5). ### Comparison of cytokine levels between patients with SLE and healthy controls Compared with controls, patients with SLE had higher average levels of BAFF (1.74 vs 0.94 ng/mL, p<0.001) and MCP-1 (137.42 vs 98.22 pg/mL, p=0.028), while having lower levels of IL-1β (26.85 vs 67.86, p<0.001) and TGF-β1 (581.74 vs 733.39, p=0.018). Within patients with SLE, smokers had higher average levels of BAFF (2.01 vs 1.61 ng/mL, p=0.034), yet lower levels of IFN-γ (39.37 vs 82.98 pg/mL, p=0.002), TNF-α (35.41 vs 58.37 pg/mL, p=0.024) and MIP-1β (509.26 vs 625.65 pg/mL, p=0.022). Compared with controls, patients with SLE who smoked had higher levels of BAFF (2.01 vs 0.94 ng/mL, p<0.001) and MCP-1 (118.9 vs 98.22 pg/mL, p=0.031), and lower IL-1β (21.85 vs 67.86 pg/mL, p=0.006) but equivalent IFN-γ levels (39.37 vs 53.88, p=0.064) (table 2). View this table: [Table 2](http://lupus.bmj.com/content/8/1/e000537/T2) Table 2 Cytokine levels in patients with SLE and 31 healthy controls (reference levels) at the research visit, compared across smoking status ### Association of smoking with cytokine levels Smoking correlated with increased BAFF levels (Rs 0.20, p=0.043) leading to 27% and 28% increased BAFF levels after adjusting for age and sex (95% CI 3% to 57%; p=0.023) and clinical covariates (model 1) (95% CI 5% to 57%, p=0.015), respectively. Smoking inversely correlated with IFN-γ levels (Rs −0.32, p=0.001) and a 55% decrease IFN-γ in both the age-adjusted and sex-adjusted (95% CI −71% to −27%, p=0.001) and clinical covariate adjusted models (95% CI −72% to −28%; p=0.001). Smoking inversely associated with TNF-α and MIP-1β levels in the univariate, age-adjusted and sex-adjusted model, and model 1. Smoking inversely correlated with IL-4 (Rs −0.25, p=0.015), but was not in the adjusted models. Smoking associated with reduced levels of IL-1β in the age-adjusted and sex-adjusted model only. Smoking associated with decreased IL-12 and IL-17 in the age-adjusted and sex-adjusted model and model 1, but not model 2 (table 3). View this table: [Table 3](http://lupus.bmj.com/content/8/1/e000537/T3) Table 3 Univariate and multivariate association between smoking and cytokine levels in patients with SLE ### Analysis further adjusting for anti-inflammatory or regulatory cytokines With further adjustment for anti-inflammatory cytokines (IL-4, IL-10 and TGF-β1) and regulatory cytokine IL-1β (model 2), smoking increased BAFF levels by 27% (95% CI 6% to 48%; p=0.014) and decreased IFN-γ levels by 42% (95% CI −79% to −5%; p=0.026); however, smoking no longer associated with changes in IL-12, IL-17, TNF-α or MIP-1β (which were dependent on IL-4 levels) (table 3). ## Discussion This cross-sectional cohort study demonstrates that in patients with SLE, current smoking associates with increased BAFF levels and decreased IFN-γ levels. This was on a background of current smokers having increased joint manifestations necessitating higher NSAID usage, migraine, Raynaud’s phenomenon and increased GDA scores. Current smokers had accrued more malar rash and mucosal ulcers over time (ACR97), but not skin damage (OR 3.82, p=0281). Having ever smoked was associated with increased odds of organ damage and cancer development. However, current smokers and non-smokers were similar with respect to the remaining ACR97 classification criteria items and scores, cross-sectional SLEDAI-2K scores, damage accrual (SDI >0, 59.6%), comorbidity and other medication requirement. The harmful effects of cigarette smoking are well established in the general population, and the more recent phenomenon of smoking e-cigarettes (vaping) is gradually being shown to equally so.31 The 35.4% of patients with SLE smoking and the 25 (39.1%) ex-smokers aligned with Norwegian national survey data on smoking prevalence and cessation at the time of the study.32 Compared with other studies of patients with SLE, smoking prevalence was equivalent to studies based in the USA (32% current smokers) and Denmark (51% ever-smoked), but higher than in Canada (14% current smokers) and Brazil (8% current smokers).7 10 11 21 Similarly, smoking intensity (cigarettes per day and pack-years) in our cohort was equivalent to those who reported it.7 11 21 Despite the Norwegian effort to promote smoking cessation, the prevalence of smoking is currently around 20%, and it is not expected to fall below 10% until 2029,32 which indicates that an opportunity still exists for clinicians to encourage smoking cessation in their patients. Our cohort was similar to other studies about the impact of smoking on SLE, with respect to participants’ age (mean 48 years old),21 disease duration (an average of 10 years)21 and sex distribution.7 11 21 22 The higher odds of prevalent (ACR97) mucosal ulcers in our cohort add to the established link between current smoking and (active) mucocutaneous features in SLE.21 In spite of the small numbers, we showed a signal of more skin damage in current smokers (OR 3.82; p>0.05), although similar for the over-representation of cumulative cutaneous features in smokers. Our findings differed to others who showed that having ever smoked7 or having had a >20 pack-year history11 increased the odds of discoid lesions and photosensitivity. The increased odds of mucosal rather than discoid lesions and photosensitivity may result from differences in the ultraviolet (UV) light exposure at each research setting,33 with Tromso (Northern Norway) having lower peak UV (~4) compared with the Canadian and Danish studies (~6 UV index).7 11 We showed that having ever smoked rather than current smoking alone resulted in proportionally more damage accrual and malignancy. This aligned with Bourré-Tessier *et al* who reported that smoking associated with lung cancer in patients with SLE.7 Taken together, patients with SLE should be advised to cease smoking to reduce the odds of mucocutaneous disease flares, damage accrual and cancer development. Similar to other cross-sectional studies, smoking exhibited little influence on total SLEDAI score, rather we showed increased odds of arthritis, migraine and vasculitis (Raynaud’s phenomenon).11 21 In contrast, other studies showed that smoking increased average SLEDAI scores,21 increased cutaneous-SLE disease activity,34 and either slowed or prevented the reduction of SLEDAI scores after treatment with belimumab.35 Herein, the increased odds of arthritis in patients with SLE who smoked contradicted the inverse association between a smoking history of >20 pack-years and with non-erosive arthritis (OR=0.45) reported by Leffers *et al*.11 However, in the context of the lower IFN-γ levels shown herein, our data align with murine models, which showed that smoking can cause a collagen-induced arthritis.36 37 Patients with SLE who smoked also had more migraine, headache and Raynaud’s phenomenon (disproportionately affecting men). This fits with the increased frequency of headache and vasculitis in active smokers, and the increased odds of neurological disorders in those with smoking history of >20 pack-years reported elsewhere.10 11 21 38 Patients with SLE who smoked also had increased patient and physician GDA scores (0–10 mm VAS). Yet, similar to Golder *et al*, we found that smoking status (current or ex-smoking) had no influence on LLDAS-30, LLDAS-50 or LLDAS-70 scores (all p>0.30).39 Thus, in patients with SLE, we suspect that the harmful effects of smoking to health, systemic inflammation and disease activity are not entirely captured by the SLEDAI-2K,23 which may mean that the impact of smoking is potentially obscured in current treat-to-target indices.39 For similar medication profiles, current smokers trended towards a reduction in the prevalence (OR 0.52, p=0.125) and severity of the typical cytopenias seen in SLE, and current smokers had increased levels of haemoglobin and MCV.40 41 This reflects the increased systemic inflammatory state brought about by smoking-induced damage to the lungs and circulation of smoking by-products throughout the body.40 41 In spite of the increased systemic inflammatory state, smoking did not influence the proportion with active hypocomplementemia and autoantibody production, which is consistent with the literature.4 10 42 43 Thus, clinicians should advise their patients that smoking causes acute systemic damage to the body, beyond what is captured within the routine clinical assessment (SLEDAI-2K) or target-to-treat indices of SLE. The increased (27%) serum BAFF levels in patients with SLE who smoked confirmed the 8.7% and 24% increase seen in (ANA−/ANA+) smokers in the Nurse’s Health Study (n=1177), respectively.5 Smoking has been shown to increase serum BAFF levels in the absence of autoimmune disease.5 44 The overexpression of BAFF in SLE is hypothesised to be driven by localised inflammation or B cell activation resulting from inherent immune dysregulation in SLE.30 45 BAFF levels have also been associated with disease activity46 and organ damage47 48 in some but not all cohort studies.45 Yet, the failure of monoclonal anti-BAFF therapy to dampen disease activity49 suggests that aberrant BAFF production is a feature rather than a direct cause of lupus disease activity.45 Furthermore, smoking has been shown to reduce the efficacy of belimumab (BAFF inhibition)35 and antimalarial agents in the management of SLE.50 Taken together, the independent association between smoking and BAFF levels, on a background of increased physician GDA score arthritis and vasculopathies, suggests that smoking cessation should be a priority in the management of SLE to reduce systemic inflammation and disease activity, which would help to reduce serum BAFF levels.30 Innate antiviral immune competence is a major function of many type I IFNs, which along with IL-12, IL-15 and IL-18, can induce type II IFN (IFN-γ) production.51 Dysregulation of type I IFN, and potentially type II IFN (IFN-γ), can lead to uncontrolled virus replication coupled with uncontrolled inflammation which causes tissue damage.51 IFN-γ in inflammatory and autoimmune pathologies has bidirectional immunoregulatory effects, which often moderate disease severity, and maintains antiviral and microbial defences, among other features.52–56 We showed increased IFN-γ levels in non-smoking patients with SLE compared with healthy controls (82.98 vs 53.88 pg/mL, p<0.001).18 57 However, patients with SLE who smoked had IFN-γ levels similar to and trending lower than those of healthy controls (39.37 vs 53.88 pg/mL, p=0.064). The suppression of IFN-γ levels within patients with SLE who smoked (model 2) aligns with the literature about smoking-induced suppression of type I and II IFN cytokines.58–62 With respect to IFNs, smoking can cause phosphorylation-dependent downregulation of IFN-1A receptor (IFN-1AR) by reactive oxygen species (ROS),63 which, although unproven as yet, may result from exposure to the major phenolic components of cigarette tar, hydroquinone and catechol.27 Additionally, the phosphorylation-dependent downregulation of IFN-1AR by ROS was ameliorated by the antioxidant N-acetylcysteine (NAC).63 Interestingly, in patients with SLE, disease activity was shown to improve within 1 month of administration of NAC (2.4–4.8 g/day),64 which suggests that perhaps, NAC supplementation may be beneficial in patients with SLE,65 especially for those struggling with smoking cessation.66 Nicotine has been recently cited as having beneficial proinflammatory and anti-inflammatory effects67; and pertinent to SLE, nicotine was shown to augment TGF-β levels and increase expression of FoxP3 in mice.68 However, in our study, TGF-β1 levels were lowest in patients with SLE who smoked. Thus, while nicotine replacement therapy has its use in smoking cessation, its psychoactive properties (addictive), vasoconstrictive properties (perhaps responsible for the increased odds of headache and Raynaud’s phenomenon seen herein) and the significantly reduced levels of TGF-β1 levels of current smokers make its potential use as an immune modulator unlikely.69 Ultimately, the ability of NAC supplementation and nicotine to influence systemic and immune-mediated inflammation requires further study in patients with SLE. The clinical phenotype of patients with SLE who smoked aligns with murine model data, in which, the absence of IFN-γ had a significant reduction in serum immunoglobulins, ANA and anti-dsDNA seropositivity, renal immune deposits and proteinuria.38 Similarly, in murine models of collagen-induced arthritis, IFN-γ was protective against arthritis by inhibiting the development of Th17 cytokines.36 37 Taken together, smoking in SLE depletes IFN-γ to levels similar to or beyond those of healthy controls, with the cost of more arthritis, migraine/headaches and vasculitis, which necessitates increased NSAID use; and while we lacked the data, we suspect that smoking would have a higher prevalence of infection or viral reactivation as well. Future research could investigate the scope for NAC supplementation to help prevent a rebounding effect of type I and II (IFN-γ) IFNs to aberrant levels in patients undergoing smoking cessation.18 70 Furthermore, use of type I or II IFN inhibitors in patients with SLE who smoke would likely be met with poorer treatment response and further increase the development and severity of viral infection or reactivation.70 Levels of IL-1β are reported to be higher in patients with SLE relative to controls, especially with higher disease activity17 71 72; and lower levels of IL-1β have been shown in well-managed,17 quiescent72 and now inversely associated with smoking in patients with SLE (age-adjusted and sex-adjusted model). Our data and others indicate that in patients with SLE, the by-products of smoking can reduce the levels of conventionally proinflammatory (TNF-α, IL-12, IL-17 and MIP-1β) and anti-inflammatory (IL-4) cytokines.27 73 While the depletion of these cytokines may not be significant enough to cause immune-mediated pathology, in the context of a reduced regulatory capacity of IL-1β,30 this may increase the susceptibility of patients with SLE to infections, atherosclerosis, cardiovascular disease and cancer development.9 74–77 Thus, we speculate that smoking cessation may help normalise levels of IL-1β in patients with SLE, which would help to inhibit undesirable immune responses, possibly by increasing the Treg functionality via Th2 cytokines, and give protection against comorbidity. The strength of this study is the availability of a large range of disease characteristics in all the patients, the inclusion of longitudinal organ damage data, and the adjustment for a range of cytokines involved in regulating or exacerbating SLE. Some of the limitations of this study lie in the fact that our patients were all of Northern European descent and were mostly in a state of low disease activity, such that results cannot be extrapolated to cohorts with a different genetic or clinical makeup. The smoking exposure was captured prospectively since SLE diagnosis (with the question ‘Have you smoked earlier or do you smoke daily?’) as well as at the research visit, meaning that the details of ex-smokers’ cigarette consumption, including ‘cigarettes smoked per day’, ‘years of continuous smoking’ and the ‘years since quit smoking’ to determine pack-years, was defined retrospectively by self-report. We lacked complete data on socioeconomic status and did not collect data on alcohol consumption, although we suspect these exposures would have minimal effect on the adjusted associations with cytokine levels. While we demonstrated an independent association between smoking and IFN-γ, we did not have data on other type I IFNs, thus we cannot determine whether smoking directly influenced IFN-γ levels via type I IFNs or other cytokines, such as IL-18, which can stimulate the production of IFN-γ. Finally, our results were based on clinical and serological findings and, therefore, cannot confirm the cellular source or causation of effects of smoking on BAFF, IFN-γ and IL-1β, for which further experimental studies will be needed. ## Conclusion Smoking independently associated with increased serum BAFF and decreased IFN-γ in patients with SLE. Smoking status associated with a clinical phenotype, which included mucocutaneous features, arthritis, migraine/lupus headaches and secondary Raynaud’s phenomenon, and necessitated increased use of NSAIDs. Smoking cessation should be encouraged in SLE with the aim to reduce systemic inflammation (BAFF and physician GDA), arthritis, vasospasticity, cutaneous features and NSAID requirement, while improving medication efficacy (hydroxychloroquine) and innate immune competence (IFN-γ). ## Data availability statement All data relevant to the study are included in the article or uploaded as supplemental information. ## Ethics statements ### Patient consent for publication Not required. ### Ethics approval Patients provided written informed consent for this study, which was approved by the regional ethics committee (REK: 2015/1400). ## Acknowledgments Andrea Becker-Merok and Kristen Nilsen. ## Footnotes * Contributors All authors have contributed to the study design and writing of this manuscript. WDR is responsible (guarantor) of the overall content. WDR conducted the statistical analysis and assures the integrity and accuracy of the data presented in this study. * Funding This work was supported by an unrestricted grant from the Arthritis and Osteoporosis Foundation of Western Australia (JN). WR received a PhD scholarship in Memory of John Donald Stewart, the Gabrielle Vitale Memorial Fund Grant from the Lupus Group of Western Australia, and the Australian Rheumatology Association (Western Australia Branch) Research & Training Scholarship. * Competing interests None declared. * Provenance and peer review Not commissioned; externally peer reviewed. * Supplemental material This content has been supplied by the author(s). It has not been vetted by BMJ Publishing Group Limited (BMJ) and may not have been peer-reviewed. Any opinions or recommendations discussed are solely those of the author(s) and are not endorsed by BMJ. BMJ disclaims all liability and responsibility arising from any reliance placed on the content. Where the content includes any translated material, BMJ does not warrant the accuracy and reliability of the translations (including but not limited to local regulations, clinical guidelines, terminology, drug names and drug dosages), and is not responsible for any error and/or omissions arising from translation and adaptation or otherwise. * Received July 14, 2021. * Accepted October 9, 2021. * © Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ. [http://creativecommons.org/licenses/by-nc/4.0/](http://creativecommons.org/licenses/by-nc/4.0/) This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See: [http://creativecommons.org/licenses/by-nc/4.0/](http://creativecommons.org/licenses/by-nc/4.0/). ## References 1. Kaul A, Gordon C, Crow MK, et al. Systemic lupus erythematosus. Nat Rev Dis Primers 2016;2:16039.[doi:10.1038/nrdp.2016.39](http://dx.doi.org/10.1038/nrdp.2016.39)pmid:http://www.ncbi.nlm.nih.gov/pubmed/27306639 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 2. Tsokos GC, Lo MS, Costa Reis P, et al. New insights into the immunopathogenesis of systemic lupus erythematosus. Nat Rev Rheumatol 2016;12:716–30.[doi:10.1038/nrrheum.2016.186](http://dx.doi.org/10.1038/nrrheum.2016.186)pmid:http://www.ncbi.nlm.nih.gov/pubmed/27872476 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1038/nrrheum.2016.186&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=27872476&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 3. Pedersen HL, Horvei KD, Thiyagarajan D, et al. Murine and human lupus nephritis: pathogenic mechanisms and theoretical strategies for therapy. Semin Nephrol 2015;35:427–38.[doi:10.1016/j.semnephrol.2015.08.004](http://dx.doi.org/10.1016/j.semnephrol.2015.08.004)pmid:http://www.ncbi.nlm.nih.gov/pubmed/26573545 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 4. Parisis D, Bernier C, Chasset F, et al. Impact of tobacco smoking upon disease risk, activity and therapeutic response in systemic lupus erythematosus: a systematic review and meta-analysis. Autoimmun Rev 2019;18:102393. [doi:10.1016/j.autrev.2019.102393](http://dx.doi.org/10.1016/j.autrev.2019.102393)pmid:http://www.ncbi.nlm.nih.gov/pubmed/31520802 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 5. Hahn J, Leatherwood C, Malspeis S, et al. Associations between smoking and systemic lupus erythematosus-related cytokines and chemokines among US female nurses. Arthritis Care Res 2020. doi:[doi:10.1002/acr.24370](http://dx.doi.org/10.1002/acr.24370). [Epub ahead of print: 03 Jul 2020].pmid:http://www.ncbi.nlm.nih.gov/pubmed/32619290 6. Barbhaiya M, Tedeschi SK, Lu B, et al. Cigarette smoking and the risk of systemic lupus erythematosus, overall and by anti-double stranded DNA antibody subtype, in the nurses' health study cohorts. Ann Rheum Dis 2018;77:196–202.[doi:10.1136/annrheumdis-2017-211675](http://dx.doi.org/10.1136/annrheumdis-2017-211675)pmid:http://www.ncbi.nlm.nih.gov/pubmed/28988206 [Abstract/FREE Full Text](http://lupus.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6ImFubnJoZXVtZGlzIjtzOjU6InJlc2lkIjtzOjg6Ijc3LzIvMTk2IjtzOjQ6ImF0b20iO3M6Mjk6Ii9sdXB1c3NjaW1lZC84LzEvZTAwMDUzNy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 7. Bourré-Tessier J, Peschken CA, Bernatsky S, et al. Association of smoking with cutaneous manifestations in systemic lupus erythematosus. Arthritis Care Res 2013;65:1275–80.[doi:10.1002/acr.21966](http://dx.doi.org/10.1002/acr.21966)pmid:http://www.ncbi.nlm.nih.gov/pubmed/23401335 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1002/acr.21966&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 8. Ward MM, Studenski S. Clinical prognostic factors in lupus nephritis. The importance of hypertension and smoking. Arch Intern Med 1992;152:2082–8.pmid:http://www.ncbi.nlm.nih.gov/pubmed/1417382 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1001/archinte.1992.00400220098017&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=1417382&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=A1992JT16300017&link_type=ISI) 9. McKown T, Schletzbaum M, Unnithan R, et al. The effect of smoking on cumulative damage in systemic lupus erythematosus: an incident cohort study. Lupus 2021;30:620–9.[doi:10.1177/0961203320988603](http://dx.doi.org/10.1177/0961203320988603)pmid:http://www.ncbi.nlm.nih.gov/pubmed/33470148 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 10. Montes RA, Mocarzel LO, Lanzieri PG, et al. Smoking and its association with morbidity in systemic lupus erythematosus evaluated by the systemic lupus international collaborating Clinics/American College of rheumatology damage index: preliminary data and systematic review. Arthritis Rheumatol 2016;68:441–8.[doi:10.1002/art.39427](http://dx.doi.org/10.1002/art.39427)pmid:http://www.ncbi.nlm.nih.gov/pubmed/26359794 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 11. Leffers HCB, Troldborg A, Voss A, et al. Smoking associates with distinct clinical phenotypes in patients with systemic lupus erythematosus: a nationwide Danish cross-sectional study. Lupus Sci Med 2021;8:e000474.[doi:10.1136/lupus-2021-000474](http://dx.doi.org/10.1136/lupus-2021-000474)pmid:http://www.ncbi.nlm.nih.gov/pubmed/33811110 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1136/lupus-2021-000474&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=33811110&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 12. Yamaguchi NH, Smoking YNH. Smoking, immunity, and DNA damage. Transl Lung Cancer Res 2019;8:S3–6.[doi:10.21037/tlcr.2019.03.02](http://dx.doi.org/10.21037/tlcr.2019.03.02)pmid:http://www.ncbi.nlm.nih.gov/pubmed/31211100 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 13. Gonzalez-Rubio J, Navarro-Lopez C, Lopez-Najera E, et al. Cytokine release syndrome (CRS) and nicotine in COVID-19 patients: trying to calm the storm. Front Immunol 2020;11:1359. [doi:10.3389/fimmu.2020.01359](http://dx.doi.org/10.3389/fimmu.2020.01359)pmid:http://www.ncbi.nlm.nih.gov/pubmed/32595653 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.3389/fimmu.2020.01359&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 14. Daloee MH, Avan A, Mirhafez SR, et al. Impact of cigarette smoking on serum pro- and anti-inflammatory cytokines and growth factors. Am J Mens Health 2017;11:1169–73.[doi:10.1177/1557988315601724](http://dx.doi.org/10.1177/1557988315601724)pmid:http://www.ncbi.nlm.nih.gov/pubmed/26345402 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 15. Olsson P, Skogstrand K, Nilsson A, et al. Smoking, disease characteristics and serum cytokine levels in patients with primary Sjögren's syndrome. Rheumatol Int 2018;38:1503–10.[doi:10.1007/s00296-018-4063-8](http://dx.doi.org/10.1007/s00296-018-4063-8)pmid:http://www.ncbi.nlm.nih.gov/pubmed/29846789 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 16. Möckel T, Basta F, Weinmann-Menke J, et al. B cell activating factor (BAFF): structure, functions, autoimmunity and clinical implications in systemic lupus erythematosus (SLE). Autoimmun Rev 2021;20:102736. [doi:10.1016/j.autrev.2020.102736](http://dx.doi.org/10.1016/j.autrev.2020.102736)pmid:http://www.ncbi.nlm.nih.gov/pubmed/33333233 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 17. Becker-Merok A, Eilertsen Gro Østli, Nossent JC. Levels of transforming growth factor-beta are low in systemic lupus erythematosus patients with active disease. J Rheumatol 2010;37:2039–45.[doi:10.3899/jrheum.100180](http://dx.doi.org/10.3899/jrheum.100180)pmid:http://www.ncbi.nlm.nih.gov/pubmed/20682675 [Abstract/FREE Full Text](http://lupus.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoianJoZXVtIjtzOjU6InJlc2lkIjtzOjEwOiIzNy8xMC8yMDM5IjtzOjQ6ImF0b20iO3M6Mjk6Ii9sdXB1c3NjaW1lZC84LzEvZTAwMDUzNy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 18. Rönnblom L, Alm GV. Systemic lupus erythematosus and the type I interferon system. Arthritis Res Ther 2003;5:68–75.[doi:10.1186/ar625](http://dx.doi.org/10.1186/ar625)pmid:http://www.ncbi.nlm.nih.gov/pubmed/12718746 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=12718746&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=000181814100004&link_type=ISI) 19. Tan EM, Cohen AS, Fries JF, et al. The 1982 revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum 1982;25:1271–7.[doi:10.1002/art.1780251101](http://dx.doi.org/10.1002/art.1780251101)pmid:http://www.ncbi.nlm.nih.gov/pubmed/7138600 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1002/art.1780251101&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=7138600&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=A1982PQ29300001&link_type=ISI) 20. Hochberg MC. Updating the American College of rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum 1997;40:1725. [doi:10.1002/art.1780400928](http://dx.doi.org/10.1002/art.1780400928)pmid:http://www.ncbi.nlm.nih.gov/pubmed/9324032 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1002/art.1780400928&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=9324032&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=A1997XY53900028&link_type=ISI) 21. Ghaussy NO, Sibbitt W, Bankhurst AD, et al. Cigarette smoking and disease activity in systemic lupus erythematosus. J Rheumatol 2003;30:1215.pmid:http://www.ncbi.nlm.nih.gov/pubmed/12784392 [Abstract/FREE Full Text](http://lupus.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoianJoZXVtIjtzOjU6InJlc2lkIjtzOjk6IjMwLzYvMTIxNSI7czo0OiJhdG9tIjtzOjI5OiIvbHVwdXNzY2ltZWQvOC8xL2UwMDA1MzcuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 22. Ekblom-Kullberg S, Kautiainen H, Alha P, et al. Smoking, disease activity, permanent damage and dsDNA autoantibody production in patients with systemic lupus erythematosus. Rheumatol Int 2014;34:341–5.[doi:10.1007/s00296-013-2889-7](http://dx.doi.org/10.1007/s00296-013-2889-7) [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1007/s00296-013-2889-7&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=24170320&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 23. Gladman DD, Ibañez D, Urowitz MB. Systemic lupus erythematosus disease activity index 2000. J Rheumatol 2002;29:288–91.pmid:http://www.ncbi.nlm.nih.gov/pubmed/11838846 [Abstract/FREE Full Text](http://lupus.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoianJoZXVtIjtzOjU6InJlc2lkIjtzOjg6IjI5LzIvMjg4IjtzOjQ6ImF0b20iO3M6Mjk6Ii9sdXB1c3NjaW1lZC84LzEvZTAwMDUzNy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 24. Franklyn K, Lau CS, Navarra SV, et al. Definition and initial validation of a lupus low disease activity state (LLDAS). Ann Rheum Dis 2016;75:1615–21.[doi:10.1136/annrheumdis-2015-207726](http://dx.doi.org/10.1136/annrheumdis-2015-207726)pmid:http://www.ncbi.nlm.nih.gov/pubmed/26458737 [Abstract/FREE Full Text](http://lupus.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6ImFubnJoZXVtZGlzIjtzOjU6InJlc2lkIjtzOjk6Ijc1LzkvMTYxNSI7czo0OiJhdG9tIjtzOjI5OiIvbHVwdXNzY2ltZWQvOC8xL2UwMDA1MzcuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 25. Sharma C, Raymond W, Eilertsen G, et al. Association of achieving lupus low disease activity state fifty percent of the time with both reduced damage Accrual and mortality in patients with systemic lupus erythematosus. Arthritis Care Res 2020;72:447–51.[doi:10.1002/acr.23867](http://dx.doi.org/10.1002/acr.23867)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30821926 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 26. Gladman D, Ginzler E, Goldsmith C, et al. The development and initial validation of the systemic lupus international collaborating Clinics/American College of rheumatology damage index for systemic lupus erythematosus. Arthritis Rheum 1996;39:363–9.[doi:10.1002/art.1780390303](http://dx.doi.org/10.1002/art.1780390303)pmid:http://www.ncbi.nlm.nih.gov/pubmed/8607884 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1002/art.1780390303&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=8607884&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=A1996TZ59900002&link_type=ISI) 27. Ouyang Y, Virasch N, Hao P, et al. Suppression of human IL-1beta, IL-2, IFN-gamma, and TNF-alpha production by cigarette smoke extracts. J Allergy Clin Immunol 2000;106:280–7.[doi:10.1067/mai.2000.107751](http://dx.doi.org/10.1067/mai.2000.107751)pmid:http://www.ncbi.nlm.nih.gov/pubmed/10932071 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1067/mai.2000.107751&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=10932071&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=000088708100010&link_type=ISI) 28. Higaki M, Wada H, Mikura S, et al. Interleukin-10 modulates pulmonary neutrophilic inflammation induced by cigarette smoke exposure. Exp Lung Res 2015;41:525–34.[doi:10.3109/01902148.2015.1096315](http://dx.doi.org/10.3109/01902148.2015.1096315)pmid:http://www.ncbi.nlm.nih.gov/pubmed/26651880 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 29. Qiu F, Liang C-L, Liu H. Impacts of cigarette smoking on immune responsiveness: up and Down or upside down? Oncotarget 2016;8. 30. Raymond WD, Eilertsen Gro Østli, Nossent J. Principal component analysis reveals disconnect between regulatory cytokines and disease activity in systemic lupus erythematosus. Cytokine 2019;114:67–73.[doi:10.1016/j.cyto.2018.10.013](http://dx.doi.org/10.1016/j.cyto.2018.10.013)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30551949 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 31. Wang Q, Sundar IK, Li D, et al. E-cigarette-induced pulmonary inflammation and dysregulated repair are mediated by nAChR α7 receptor: role of nAChR α7 in SARS-CoV-2 Covid-19 ACE2 receptor regulation. Respir Res 2020;21:154. [doi:10.1186/s12931-020-01396-y](http://dx.doi.org/10.1186/s12931-020-01396-y)pmid:http://www.ncbi.nlm.nih.gov/pubmed/32552811 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1186/s12931-020-01396-y&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 32. Gartner CE, Lund KE, Barendregt JJ, et al. Projecting the future smoking prevalence in Norway. Eur J Public Health 2016;27:ckw180–44.[doi:10.1093/eurpub/ckw180](http://dx.doi.org/10.1093/eurpub/ckw180) 33. Kim A, Chong BF. Photosensitivity in cutaneous lupus erythematosus. Photodermatol Photoimmunol Photomed 2013;29:4–11.[doi:10.1111/phpp.12018](http://dx.doi.org/10.1111/phpp.12018)pmid:http://www.ncbi.nlm.nih.gov/pubmed/23281691 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1111/phpp.12018&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=23281691&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 34. Ezra N, Jorizzo J. Hydroxychloroquine and smoking in patients with cutaneous lupus erythematosus. Clin Exp Dermatol 2012;37:327–34.[doi:10.1111/j.1365-2230.2011.04266.x](http://dx.doi.org/10.1111/j.1365-2230.2011.04266.x) [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1111/j.1365-2230.2011.04266.x&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=22582908&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 35. Parodis I, Sjöwall C, Jönsen A, et al. Smoking and pre-existing organ damage reduce the efficacy of belimumab in systemic lupus erythematosus. Autoimmun Rev 2017;16:343–51.[doi:10.1016/j.autrev.2017.02.005](http://dx.doi.org/10.1016/j.autrev.2017.02.005)pmid:http://www.ncbi.nlm.nih.gov/pubmed/28216072 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 36. Pollard KM, Cauvi DM, Toomey CB, et al. Interferon-γ and systemic autoimmunity. Discov Med 2013;16:123–31.pmid:http://www.ncbi.nlm.nih.gov/pubmed/23998448 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=23998448&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 37. Malenica M, Prnjavorac B, Bego T, et al. Effect of cigarette smoking on haematological parameters in healthy population. Med Arch 2017;71:132–6.[doi:10.5455/medarh.2017.71.132-136](http://dx.doi.org/10.5455/medarh.2017.71.132-136)pmid:http://www.ncbi.nlm.nih.gov/pubmed/28790546 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 38. Suter LG, Murabito JM, Felson DT, et al. Smoking, alcohol consumption, and Raynaud's phenomenon in middle age. Am J Med 2007;120:264–71.[doi:10.1016/j.amjmed.2006.06.007](http://dx.doi.org/10.1016/j.amjmed.2006.06.007)pmid:http://www.ncbi.nlm.nih.gov/pubmed/17349450 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1016/j.amjmed.2006.06.007&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=17349450&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=000244756400016&link_type=ISI) 39. Golder V, Kandane-Rathnayake R, Hoi AY-B, et al. Frequency and predictors of the lupus low disease activity state in a multi-national and multi-ethnic cohort. Arthritis Res Ther 2016;18:260–60.[doi:10.1186/s13075-016-1163-2](http://dx.doi.org/10.1186/s13075-016-1163-2)pmid:http://www.ncbi.nlm.nih.gov/pubmed/27829463 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 40. Eisenga MF, Wouters HJCM, Kieneker LM, et al. Active smoking and macrocytosis in the general population: two population-based cohort studies. Am J Hematol 2019;94:E45–8.[doi:10.1002/ajh.25346](http://dx.doi.org/10.1002/ajh.25346)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30417934 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 41. Pedersen KM, Çolak Y, Ellervik C, et al. Smoking and increased white and red blood cells. Arterioscler Thromb Vasc Biol 2019;39:965–77.[doi:10.1161/ATVBAHA.118.312338](http://dx.doi.org/10.1161/ATVBAHA.118.312338)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30866659 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 42. Xu D, You X, Wang Z, et al. Chinese systemic lupus erythematosus treatment and research Group registry VI: effect of cigarette smoking on the clinical phenotype of Chinese patients with systemic lupus erythematosus. PLoS One 2015;10:e0134451. [doi:10.1371/journal.pone.0134451](http://dx.doi.org/10.1371/journal.pone.0134451)pmid:http://www.ncbi.nlm.nih.gov/pubmed/26280671 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1371/journal.pone.0134451&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=26280671&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 43. Young KA, Terrell DR, Guthridge JM, et al. Smoking is not associated with autoantibody production in systemic lupus erythematosus patients, unaffected first-degree relatives, nor healthy controls. Lupus 2014;23:360–9.[doi:10.1177/0961203314520838](http://dx.doi.org/10.1177/0961203314520838)pmid:http://www.ncbi.nlm.nih.gov/pubmed/24449338 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1177/0961203314520838&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=24449338&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=000337296000004&link_type=ISI) 44. Battaglino RA, Nguyen N, Summers M, et al. B cell-activating factor is associated with testosterone and smoking status in Non-Ambulatory men with chronic spinal cord injury. J Neurotrauma 2019;36:3332–7.[doi:10.1089/neu.2018.6221](http://dx.doi.org/10.1089/neu.2018.6221)pmid:http://www.ncbi.nlm.nih.gov/pubmed/31020912 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 45. Eilertsen Gro Ø, Van Ghelue M, Strand H, et al. Increased levels of BAFF in patients with systemic lupus erythematosus are associated with acute-phase reactants, independent of BAFF genetics: a case-control study. Rheumatology 2011;50:2197–205.[doi:10.1093/rheumatology/ker282](http://dx.doi.org/10.1093/rheumatology/ker282)pmid:http://www.ncbi.nlm.nih.gov/pubmed/21984763 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1093/rheumatology/ker282&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=21984763&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=000297368400009&link_type=ISI) 46. Howe HS, Thong BYH, Kong KO, et al. Associations of B cell-activating factor (BAFF) and anti-BAFF autoantibodies with disease activity in multi-ethnic Asian systemic lupus erythematosus patients in Singapore. Clin Exp Immunol 2017;189:298–303.[doi:10.1111/cei.12975](http://dx.doi.org/10.1111/cei.12975) 47. McCarthy EM, Lee RZ, Ní Gabhann J, et al. Elevated B lymphocyte stimulator levels are associated with increased damage in an Irish systemic lupus erythematosus cohort. Rheumatology 2013;52:1279–84.[doi:10.1093/rheumatology/ket120](http://dx.doi.org/10.1093/rheumatology/ket120)pmid:http://www.ncbi.nlm.nih.gov/pubmed/23479724 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1093/rheumatology/ket120&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=23479724&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=000321061200021&link_type=ISI) 48. Vincent FB, Kandane‐Rathnayake R, Koelmeyer R, et al. Analysis of serum B cell‐activating factor from the tumor necrosis factor family (BAFF) and its soluble receptors in systemic lupus erythematosus. Clin Transl Immunology 2019;8:e1047. [doi:10.1002/cti2.1047](http://dx.doi.org/10.1002/cti2.1047) 49. Baker KP, Edwards BM, Main SH, et al. Generation and characterization of LymphoStat-B, a human monoclonal antibody that antagonizes the bioactivities of B lymphocyte stimulator. Arthritis Rheum 2003;48:3253–65.[doi:10.1002/art.11299](http://dx.doi.org/10.1002/art.11299)pmid:http://www.ncbi.nlm.nih.gov/pubmed/14613291 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1002/art.11299&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=14613291&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=000186451100034&link_type=ISI) 50. Chasset F, Francès C, Barete S, et al. Influence of smoking on the efficacy of antimalarials in cutaneous lupus: a meta-analysis of the literature. J Am Acad Dermatol 2015;72:634–9.[doi:10.1016/j.jaad.2014.12.025](http://dx.doi.org/10.1016/j.jaad.2014.12.025)pmid:http://www.ncbi.nlm.nih.gov/pubmed/25648824 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 51. Lee AJ, Ashkar AA. The dual nature of type I and type II interferons. Front Immunol 2018;9:9. [doi:10.3389/fimmu.2018.02061](http://dx.doi.org/10.3389/fimmu.2018.02061)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30254639 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.3389/fimmu.2018.00288&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=29403493&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 52. Harris N, Buller RM, Karupiah G. Gamma interferon-induced, nitric oxide-mediated inhibition of vaccinia virus replication. J Virol 1995;69:910–5.[doi:10.1128/jvi.69.2.910-915.1995](http://dx.doi.org/10.1128/jvi.69.2.910-915.1995)pmid:http://www.ncbi.nlm.nih.gov/pubmed/7529336 [Abstract/FREE Full Text](http://lupus.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoianZpIjtzOjU6InJlc2lkIjtzOjg6IjY5LzIvOTEwIjtzOjQ6ImF0b20iO3M6Mjk6Ii9sdXB1c3NjaW1lZC84LzEvZTAwMDUzNy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 53. Buchmeier NA, Schreiber RD. Requirement of endogenous interferon-gamma production for resolution of Listeria monocytogenes infection. Proc Natl Acad Sci U S A 1985;82:7404–8.[doi:10.1073/pnas.82.21.7404](http://dx.doi.org/10.1073/pnas.82.21.7404)pmid:http://www.ncbi.nlm.nih.gov/pubmed/3933006 [Abstract/FREE Full Text](http://lupus.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMDoiODIvMjEvNzQwNCI7czo0OiJhdG9tIjtzOjI5OiIvbHVwdXNzY2ltZWQvOC8xL2UwMDA1MzcuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 54. Suzuki Y, Orellana MA, Schreiber RD, et al. Interferon-Gamma: the major mediator of resistance against Toxoplasma gondii. Science 1988;240:516–8.[doi:10.1126/science.3128869](http://dx.doi.org/10.1126/science.3128869)pmid:http://www.ncbi.nlm.nih.gov/pubmed/3128869 [Abstract/FREE Full Text](http://lupus.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIyNDAvNDg1MS81MTYiO3M6NDoiYXRvbSI7czoyOToiL2x1cHVzc2NpbWVkLzgvMS9lMDAwNTM3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 55. Murray HW, Rubin BY, Rothermel CD. Killing of intracellular Leishmania donovani by lymphokine-stimulated human mononuclear phagocytes. Evidence that interferon-gamma is the activating lymphokine. J Clin Invest 1983;72:1506–10.[doi:10.1172/JCI111107](http://dx.doi.org/10.1172/JCI111107)pmid:http://www.ncbi.nlm.nih.gov/pubmed/6415111 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1172/JCI111107&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=6415111&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=A1983RM65100035&link_type=ISI) 56. Kelchtermans H, Billiau A, Matthys P. How interferon-gamma keeps autoimmune diseases in check. Trends Immunol 2008;29:479–86.[doi:10.1016/j.it.2008.07.002](http://dx.doi.org/10.1016/j.it.2008.07.002)pmid:http://www.ncbi.nlm.nih.gov/pubmed/18775671 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1016/j.it.2008.07.002&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=18775671&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=000260445000004&link_type=ISI) 57. Munroe ME, Lu R, Zhao YD, et al. Altered type II interferon precedes autoantibody accrual and elevated type I interferon activity prior to systemic lupus erythematosus classification. Ann Rheum Dis 2016;75:2014–21.[doi:10.1136/annrheumdis-2015-208140](http://dx.doi.org/10.1136/annrheumdis-2015-208140)pmid:http://www.ncbi.nlm.nih.gov/pubmed/27088255 [Abstract/FREE Full Text](http://lupus.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6ImFubnJoZXVtZGlzIjtzOjU6InJlc2lkIjtzOjEwOiI3NS8xMS8yMDE0IjtzOjQ6ImF0b20iO3M6Mjk6Ii9sdXB1c3NjaW1lZC84LzEvZTAwMDUzNy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 58. Liu G, Arimilli S, Savage E, et al. Cigarette smoke preparations, not moist snuff, impair expression of genes involved in immune signaling and cytolytic functions. Sci Rep 2019;9:13390. [doi:10.1038/s41598-019-48822-w](http://dx.doi.org/10.1038/s41598-019-48822-w)pmid:http://www.ncbi.nlm.nih.gov/pubmed/31527707 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 59. Modestou MA, Manzel LJ, El-Mahdy S, et al. Inhibition of IFN-gamma-dependent antiviral airway epithelial defense by cigarette smoke. Respir Res 2010;11:64. [doi:10.1186/1465-9921-11-64](http://dx.doi.org/10.1186/1465-9921-11-64)pmid:http://www.ncbi.nlm.nih.gov/pubmed/20504369 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1186/1465-9921-11-64&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=20504369&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 60. Shin SS, Gallardo M, Lozada R, et al. Cigarette-Smoking intensity and interferon-gamma release assay conversion among persons who inject drugs: a cohort study. Pulm Med 2012;2012:1–7.[doi:10.1155/2012/828106](http://dx.doi.org/10.1155/2012/828106)pmid:http://www.ncbi.nlm.nih.gov/pubmed/23304497 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 61. Vassallo R, Chen L. Selective suppression of dendritic cell functions by cigarette smoke extract. Chest 2004;125:107s. [doi:10.1378/chest.125.5\_suppl.107S](http://dx.doi.org/10.1378/chest.125.5_suppl.107S)pmid:http://www.ncbi.nlm.nih.gov/pubmed/15136445 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 62. Bauer CMT, Dewitte-Orr SJ, Hornby KR, et al. Cigarette smoke suppresses type I interferon-mediated antiviral immunity in lung fibroblast and epithelial cells. J Interferon Cytokine Res 2008;28:167–79.[doi:10.1089/jir.2007.0054](http://dx.doi.org/10.1089/jir.2007.0054)pmid:http://www.ncbi.nlm.nih.gov/pubmed/18338949 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1089/jir.2007.0054&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=18338949&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=000254301900004&link_type=ISI) 63. HuangFu W-C, Liu J, Harty RN, et al. Cigarette smoking products suppress anti-viral effects of type I interferon via phosphorylation-dependent downregulation of its receptor. FEBS Lett 2008;582:3206–10.[doi:10.1016/j.febslet.2008.08.013](http://dx.doi.org/10.1016/j.febslet.2008.08.013) [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1016/j.febslet.2008.08.013&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=18722370&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=000259123800008&link_type=ISI) 64. Lai Z-W, Hanczko R, Bonilla E, et al. N -acetylcysteine reduces disease activity by blocking mammalian target of rapamycin in T cells from systemic lupus erythematosus patients: A randomized, double-blind, placebo-controlled trial. Arthritis & Rheumatism 2012;64:2937–46.[doi:10.1002/art.34502](http://dx.doi.org/10.1002/art.34502) [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1002/art.34502&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=22549432&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=000308035100014&link_type=ISI) 65. Perl A. Oxidative stress in the pathology and treatment of systemic lupus erythematosus. Nat Rev Rheumatol 2013;9:674–86.[doi:10.1038/nrrheum.2013.147](http://dx.doi.org/10.1038/nrrheum.2013.147)pmid:http://www.ncbi.nlm.nih.gov/pubmed/24100461 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1038/nrrheum.2013.147&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=24100461&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 66. Islam MA, Khandker SS, Kotyla PJ, et al. Immunomodulatory effects of diet and nutrients in systemic lupus erythematosus (SLE): a systematic review. Front Immunol 2020;11.[doi:10.3389/fimmu.2020.01477](http://dx.doi.org/10.3389/fimmu.2020.01477) 67. Piao W-H, Campagnolo D, Dayao C, et al. Nicotine and inflammatory neurological disorders. Acta Pharmacol Sin 2009;30:715–22.[doi:10.1038/aps.2009.67](http://dx.doi.org/10.1038/aps.2009.67)pmid:http://www.ncbi.nlm.nih.gov/pubmed/19448649 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1038/aps.2009.67&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=19448649&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=000266916900008&link_type=ISI) 68. Shi F-D, Piao W-H, Kuo Y-P, et al. Nicotinic attenuation of central nervous system inflammation and autoimmunity. J Immunol 2009;182:1730–9.[doi:10.4049/jimmunol.182.3.1730](http://dx.doi.org/10.4049/jimmunol.182.3.1730)pmid:http://www.ncbi.nlm.nih.gov/pubmed/19155522 [Abstract/FREE Full Text](http://lupus.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTA6IjE4Mi8zLzE3MzAiO3M6NDoiYXRvbSI7czoyOToiL2x1cHVzc2NpbWVkLzgvMS9lMDAwNTM3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 69. Liberman K, Van Schuerbeek P, Herremans S, et al. The effect of nicotine patches on craving in the brain: a functional MRI study on heavy smokers. Medicine 2018;97:e12415–e15.[doi:10.1097/MD.0000000000012415](http://dx.doi.org/10.1097/MD.0000000000012415)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30278517 [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 70. Morand EF, Furie R, Tanaka Y, et al. Trial of Anifrolumab in active systemic lupus erythematosus. N Engl J Med 2020;382:211–21.[doi:10.1056/NEJMoa1912196](http://dx.doi.org/10.1056/NEJMoa1912196)pmid:http://www.ncbi.nlm.nih.gov/pubmed/31851795 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1056/NEJMoa1912196&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 71. McCarthy EM, Smith S, Lee RZ, et al. The association of cytokines with disease activity and damage scores in systemic lupus erythematosus patients. Rheumatology 2014;53:1586–94.[doi:10.1093/rheumatology/ket428](http://dx.doi.org/10.1093/rheumatology/ket428)pmid:http://www.ncbi.nlm.nih.gov/pubmed/24706988 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1093/rheumatology/ket428&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=24706988&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 72. Umare V, Pradhan V, Nadkar M, et al. Effect of proinflammatory cytokines (IL-6, TNF-α, and IL-1β) on clinical manifestations in Indian SLE patients. Mediators Inflamm 2014;2014:1–8.[doi:10.1155/2014/385297](http://dx.doi.org/10.1155/2014/385297)pmid:http://www.ncbi.nlm.nih.gov/pubmed/25548434 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1155/2014/319215&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=25197169&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) 73. Paludan SR. Interleukin-4 and interferon-gamma: the quintessence of a mutual antagonistic relationship. Scand J Immunol 1998;48:459–68.[doi:10.1046/j.1365-3083.1998.00435.x](http://dx.doi.org/10.1046/j.1365-3083.1998.00435.x)pmid:http://www.ncbi.nlm.nih.gov/pubmed/9822252 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1046/j.1365-3083.1998.00435.x&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=9822252&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=000076942200002&link_type=ISI) 74. Bradley JR. Tnf-Mediated inflammatory disease. J Pathol 2008;214:149–60.[doi:10.1002/path.2287](http://dx.doi.org/10.1002/path.2287)pmid:http://www.ncbi.nlm.nih.gov/pubmed/18161752 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1002/path.2287&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=18161752&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=000252597000004&link_type=ISI) 75. Maurer M, von Stebut E. Macrophage inflammatory protein-1. Int J Biochem Cell Biol 2004;36:1882–6.[doi:10.1016/j.biocel.2003.10.019](http://dx.doi.org/10.1016/j.biocel.2003.10.019)pmid:http://www.ncbi.nlm.nih.gov/pubmed/15203102 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.1016/j.biocel.2003.10.019&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=15203102&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom) [Web of Science](http://lupus.bmj.com/lookup/external-ref?access_num=000222886600005&link_type=ISI) 76. Hagberg N, Berggren O, Leonard D, et al. IFN-α production by plasmacytoid dendritic cells stimulated with RNA-containing immune complexes is promoted by NK cells via MIP-1β and LFA-1. J Immunol 2011;186:5085–94.[doi:10.4049/jimmunol.1003349](http://dx.doi.org/10.4049/jimmunol.1003349)pmid:http://www.ncbi.nlm.nih.gov/pubmed/21430220 [Abstract/FREE Full Text](http://lupus.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTA6IjE4Ni85LzUwODUiO3M6NDoiYXRvbSI7czoyOToiL2x1cHVzc2NpbWVkLzgvMS9lMDAwNTM3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 77. Raymond W, Ostli-Eilertsen G, Griffiths S, et al. Il-17A levels in systemic lupus erythematosus associated with inflammatory markers and lower rates of malignancy and heart damage: evidence for a dual role. Eur J Rheumatol 2017;4:29–35.[doi:10.5152/eurjrheum.2017.16059](http://dx.doi.org/10.5152/eurjrheum.2017.16059)pmid:http://www.ncbi.nlm.nih.gov/pubmed/28293450 [CrossRef](http://lupus.bmj.com/lookup/external-ref?access_num=10.5152/eurjrheum.2017.16059&link_type=DOI) [PubMed](http://lupus.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Flupusscimed%2F8%2F1%2Fe000537.atom)