Skip to main content

Psoriasis may increase the risk of idiopathic pulmonary fibrosis: a two-sample Mendelian randomization study

Abstract

Background

Although some studies have indicated that Psoriasis could contribute to the risk of idiopathic pulmonary fibrosis (IPF), no study has reported a clear causal association between them. Our aim was to explore the potential relationship between Psoriasis and IPF using Mendelian randomization (MR) design.

Methods

To explore a causal association between Psoriasis and IPF, we used genetic instruments from the largest available genome-wide association study (GWAS) of European ancestry, including psoriasis (5314 cases, 457,619 controls) and IPF (1028 cases, 196,986 controls). Our main analyses were conducted by inverse-variance weighted (IVW) method with random-effects model, with the other complementary four analyses: weighted median method, weighted mode, multivariable MR and MR-Egger approach.

Results

The results of IVW methods demonstrated that genetically predicted psoriasis was significantly associated with higher odds of IPF, with an odds ratio (OR) of 1.09 (95%CI, 1.01–1.18; P = 0.02). Weighted median method, weighted mode and multivariable MR also demonstrated directionally similar results (P < 0.05), while the MR-Egger regression did not reveal the impact of psoriasis on IPF (OR = 1.09, 95%CI, 0.98–1.21; P = 0.11). In addition, both funnel plots and MR-Egger intercepts indicated no directional pleiotropic effects between psoriasis and IPF.

Conclusions

Our study provided potential evidence between genetically predicted psoriasis and IPF, which suggests that understanding the mutual risk factors between psoriasis and IPF can facilitate the clinical management of both diseases.

Introduction

Psoriasis is an autoimmune, chronic inflammatory diseases of skin which usually manifested as raised, well-demarcated, erythematous oval plaques with adherent silvery scales, affecting approximately 2% of the population in the world [1]. Findings from immunological and genetic studies have highlighted the crucial role of crosstalk between the innate and adaptive immune systems and the link to genetics in the development of psoriasis [2]. Specifically, keratinocytes and fibroblasts amplify inflammatory responses in psoriasis [3]. Furthermore, studies have also revealed that psoriasis patients have a distinct configuration of skin microbiota, particularly streptococcal, may be one of the triggering factors for psoriasis [4, 5]. The association of psoriasis with several systemic comorbidities, including cardiovascular disease, inflammatory bowel disease, chronic kidney disease, and chronic lung diseases like COPD, asthma has received substantial attention [6,7,8,9,10]. In the past, psoriasis and psoriatic arthritis were not considered diseases closely associated with Interstitial Lung Disease (ILD). But an increasing body of evidence suggests that pulmonary fibrosis may be more concurrent in patients with psoriasis or psoriatic arthritis than in the general population [11,12,13,14], particularly in people with severe psoriasis [15].

Idiopathic Pulmonary Fibrosis (IPF) is the most common subtype of ILD characterized by idiopathic and progressive fibrosis, a lung disease with high mortality and limited treatment options [16]. IPF is intricately linked to both genetic predisposition and recurrent environmental exposures. Dysregulated epithelial-fibroblast cross-talk plays a pivotal role, fostering an anomalous and persistent inflammatory response [17]. Recent investigations have advanced the notion that deviations in bacterial burden, diversity, and composition within the pulmonary microbiome of individuals afflicted with IPF could play a significant role in shaping disease pathogenesis and its subsequent progression [18]. Previous meta-analyses have reported microbial dysbiosis and abnormal inflammatory responses in both populations of individuals with IPF and psoriasis [19, 20]. Therefore, psoriasis may potentially be linked to an increased risk of developing IPF in theory. It was once thought that the potential correlation between the two might be attributed to the presence of a rare but recognized complication of immunosuppressants (i.e. Methotrexate, Azathioprine) [21, 22]. Existing evidence from observational studies only suggests a susceptibility of psoriasis patients to concomitant ILD. For instance, Ishikawa et al. reported 21 (4.7%) of 447 patients had the simultaneous existence of psoriasis and interstitial pneumonia [13]. Butt et al. conducted a descriptive study, they retrospectively selected 44 patients with psoriasis or psoriatic arthritis who had clinical evidence of diffuse parenchymal lung disease (DPLD, also known as ILD), where nearly one-third of patients had no prior immunosuppression, and nonspecific interstitial pneumonia (NSIP, a specific type of ILD) and unclassifiable fibrosis were seen in 24 patients (55%) and 8 patients (18%), respectively [14]. In parallel, Wu et al. and Makredes et al. conducted investigations estimating the odds ratios (ORs) for patients with psoriasis or psoriatic arthritis, revealing an elevated prevalence ratio associated with pulmonary fibrosis, ranging from 1.3 (95% confidence interval [CI], 1.1–1.5) to 1.9 (95% CI, 1.2–3.0), indicating that individuals with psoriasis may be at an increased risk of developing pulmonary fibrosis [11, 12]. Although pulmonary fibrosis is a common pathological feature in various subtypes of ILD, including IPF. There is still a lack of direct evidence linking psoriasis to IPF. In addition, both diseases above shared some common risk factors such as smoking, obesity, alcohol consumption, infections, medications, and depression [19]. And these observational studies are often limited to residual confounding and reverse causation. Therefore, the causal relationship between psoriasis and IPF remains uncertain.

Currently, Mendelian Randomization (MR) analysis has been widely used to assess the potential causal relationships between exposures and clinical outcomes [23]. Since random segregation of genetic instruments takes place before the onset of the disease, the independent assortment of genetic polymorphisms ensures the stability of MR analysis, rendering it less susceptible to environmental influences. Compared to traditional observational studies, MR analysis can overcome reverse causality, and minimize the effect of confounding factors [24]. The current study applied MR analysis to examine the causal relationship, the strength of association, and the direction of causality between psoriasis and IPF.

Materials and methods

Study design

The schematic diagram of the study design and the three key assumptions of MR are shown in Fig. 1. (A) The single nucleotide polymorphisms (SNPs) are strongly associated with psoriasis. (B) the SNPs are independent of known confounding factors associated with psoriasis. (C) The SNPs only affect idiopathic pulmonary fibrosis (IPF) through psoriasis (Fig. 1) [24].

Fig. 1
figure 1

Diagram for key assumptions of MR analyses. A SNPs are strongly associated with psoriasis; B SNPs are independent of confounders; C SNPs must only affect IPF via psoriasis. SNP single-nucleotide polymorphism

Data sources

This is a two-sample MR analysis based on summary statistics from the largest available genome-wide association studies (GWAS), including IEU openGWAS and NealELab. Both GWAS were conducted in people of European ancestry and included both males and females. The GWAS summary statistics to identify genetic risk variants for psoriasis (n = 462,933) were obtained from UK biobank study. The database included 5314 cases with a psoriasis diagnosis plus a control group of 457,619 individuals without psoriasis. Similarly, the GWAS summary statistics for IPF (n = 198,014) were obtained from the FinnGen Biobank Analysis Consortium 2021 as well as 1028 cases and 196,986 controls. The diagnosis of psoriasis and IPF was according to the ICD-10 (International Classification of diseases) criteria. Ethics approval was not required for the current analysis as all included GWAS data are publicly available and had been approved by the corresponding ethical review boards.

Selection and validation of SNPs

According to the three key assumptions of MR analysis, first, we selected a genome-wide significance level of P < 5 × 10–8. Second, a clumping algorithm with a cutoff of r2 = 0.001 and kb = 10,000 were used to avoid linkage disequilibrium (LD). When r2 > 0.001, the SNP correlated with more SNPs or with a higher P-value was deleted. Third, the F-statistic was calculated to validate the strength of individual SNPs. When F-statistics were greater than 10, SNPs were considered powerful enough to mitigate the influence of potential bias. We also conducted data-harmonization steps to ensure the effects of exposure on the outcome are consistent in terms of the effect allele and subsequent analysis.

Gene ontology and pathway enrichment analysis

To explore the biological functions and mechanism pathways for these SNPs, the risk SNPs of psoriasis patients were screened for functional enrichment. GO analysis was used to evaluate the degree of enrichment of the SNPs in biological processes, cellular components, and molecular functions. The KEGG pathway is known to grasp the metabolic processes and makes the considerable utility of genomic analysis. The P value < 0.05 was considered as a standard metric for quantifying the top listed pathways.

Statistical analysis

The primary analysis was conducted using an inverse-variance weighted (IVW) analysis under a random-effects model to combine the instrumental variable-ratio estimates of psoriasis related SNPs [25]. The weighted-median method and MR-Egger method were performed as sensitivity analyses. The weighted-median method can provide valid estimates if over 50% of information comes from valid instrumental variables (IVs) [26]. The MR-Egger method can be used to assess the horizontal pleiotropy of selected IVs [27]. We examined the heterogeneity of the ratio estimators using Cochrane’s Q-value and the MR–Egger intercept among selected IVs. Additionally, a leave one-out sensitivity analysis was conducted to determine whether the overall estimates were disproportionately affected by an individual SNP. All analyses were performed by “TwoSampleMR” packages in R version 4.2.2.

Results

Psoriasis related SNPs

In this study, we extracted twenty independent genome-wide significant (r2 < 0.001, P < 5 × 10 − 8) SNPs as IVs from the GWAS on psoriasis based on a total sample of 5314 psoriasis controls and 457,619 cases. The summary sample was almost entirely of European ancestry. All SNPs used in the MR analysis were robust with F-statistics greater than ten (Table 1). And scatter plot demonstrated the effect of each SNP locus on IPF is shown in Fig. 2. The F statistics of our SNPs were entirely > 30 (range 31.03–3089.49), which indicated that there was not a weak IVs bias for the results.

Table 1 The detailed information of 20 SNPs Association with psoriasis
Fig. 2
figure 2

Scatter plot to visualize causal effect of psoriasis on the risk of IPF. The slope of the straight line indicates the magnitude of the causal association. IVW indicates inverse-variance weighted, MR Mendelian randomization

Effect of psoriasis on IPF risk

IVW, weighted median method and MR-Egger regression were used to estimate causal association between psoriasis and the risk of IPF. The IVW analysis showed that the genetically predicted psoriasis increases the risk of IPF, with an odds ratio (OR) of 1.09 (95% confidence interval (CI) 1.01–1.18; P = 0.02). Additionally, the weighted median method (OR, 1.11 (95%CI 1.01–1.22; P = 0.03) and weighted mode (OR, 1.11 (95%CI 1.02–1.21; P = 0.03) had the similar result. MR-Egger regression (OR, 1.09 (95%CI 0.98–1.21; P = 0.11) revealed consistent estimates but of low precision (Fig. 3). In addition, the MR-Egger intercept did not significantly deviate from zero in our study (P = 0.97), suggesting no evidence of ‘horizontal pleiotropy’ or violation of the second MR assumption. Besides, to minimize the influence of common confounding factors, we conducted multi-variable adjustments in the MR analysis. Smoking was included as additional covariates in the regression model to mitigate the confounding effect of smoking on the relationship between psoriasis and idiopathic pulmonary fibrosis risk. Multivariable MR still indicated the similar result (OR, 1.09 (95%CI 1.002–1.19; P = 0.046).

Fig. 3
figure 3

Forest plot to visualize causal effect of psoriasis on the risk of IPF. IVW indicates inverse-variance weighted

Funnel plots indicated no evidence of horizontal pleiotropy (Fig. 4), as it showed that the Wald ratio of each SNP plotted was opposite to their accuracy, while asymmetry indicated directional horizontal gene pleiotropy. The Cochran Q statistics showed that the effect of SNPs included has no significant heterogeneity (Q = 19.12, P = 0.44). Furthermore, Leave-one-out analysis showed that the overall estimates were not disproportionately influenced by any individual variant (Fig. 5).

Fig. 4
figure 4

Funnel plots to visualize overall heterogeneity of Mendelian randomization (MR) estimates for the effect of psoriasis on the risk of IPF. IVW indicates inverse variance weighted

Fig. 5
figure 5

Leave-one-out plot to visualize causal effect of psoriasis on the risk of IPF when leaving one SNP out

We performed an analysis of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) using 20 SNPs in this MR study, which showed that cytokine-mediated signaling pathway was the most significant biological pathway and cytokine receptor binding was the top molecular function (Fig. 6).

Fig. 6
figure 6

Results of GO and KEGG enrichment analysis using 20 SNPs obtained from MR study. BP biological processes, CC cellular component analysis, MF molecular function analysis

Discussion

In the present two-sample MR study, we first set out to assess whether psoriasis, an autoimmune skin disorder, is causally linked to the development of IPF, a condition characterized by a median survival rate of merely 2.5–3.5 years following diagnosis. Our results revealed that the genetic predisposition of psoriasis was associated with an increased risk of IPF (OR, 1.09, 95%CI 1.01–1.18; P = 0.02) in European decent. The positive relationship between two diseases was further verified by sensitive analyses. The results provided compelling insights into the shared genetic architecture, suggesting an increased risk of IPF in psoriasis patients.

In history, psoriasis and ILD were two seemingly disparate diseases. However, as described above, emerging evidence of recent observational studies revealed that ILD were more common in patients with psoriasis than in the general population. Two studies indicated that patients with psoriasis or psoriatic arthritis had an increased prevalence ratio associated with pulmonary fibrosis ranging from 1.3 to 1.9 [11, 12]. A recent observational study in Japan by Kawamoto et.al demonstrated that the incidence of developing interstitial pneumonia in patients with psoriasis was 2%, which is significantly higher than 0.01% in natural condition [28]. Moreover, although ILD was occasionally reported in patients with psoriasis as drug-induced pneumonitis secondary to concomitant use of immunosuppressants in most cases, several cases reported simultaneous existence of psoriasis and ILD with no previous history of immunosuppressant use or biologics [13, 14, 29]. Given that IPF is the most common subtype of ILD, the association between psoriasis and IPF may be underestimated. Concordant with the findings of these observational studies, our study revealed that genetic predisposition to psoriasis was causally associated with a 9.00% increased risk of IPF.

The underlying mechanism explaining how psoriasis increases the risk of IPF remained unknown. Chronic inflammation is postulated as the central etiological factor underlying both conditions. Considerable pro-inflammatory cytokines, including tumor necrosis factor (TNF)-α, type I interferon (IFN), and notably interleukin (IL)-17/23, emanating from cutaneous lesions in psoriasis patients, elicit an aberrant immune response within the pulmonary system via the circulatory system. These same inflammatory cytokines have also been detected in individuals diagnosed with IPF [30, 31], which aligns seamlessly with the SNP instruments we have uncovered. Among the included SNPs, those situated within key loci associated with high levels of T-cell cytokines, such as rs848 within the IL-13 gene and rs11581607 within the IL-23R gene, as well as transcription factors like rs8016947 in the NF-κB gene, demonstrated notable significance in regulating immune responses and inflammation. Furthermore, other variants located in inflammatory cytokine genes, including rs12188300 (IL-12 gene), rs33980500 (TRAF3IP2 gene) and rs582757 (TNFAIP3 gene) were also found to play a role in modulating immune and inflammatory processes. Regarding the relationship between IL-12 and IPF, results are still inconsistent [32]. Some studies suggest that IL-12 may play a certain role in the pathogenesis of IPF, cause IL-12 can stimulate the production of IFN-gamma [33], and the paucity of IFN-gamma may favor the development of progressive fibrosis in IPF [34, 35]. As for the TRAF3IP2 [36] and TNFAIP3 [37] genes, some animal studies have reported pro-fibrotic responses, but a clear association with IPF has not been established. Our findings suggest that the influence of psoriasis on IPF could potentially be elucidated through microbial factors, particularly viral infections. Our GO/KEGG analysis also showed that regulation of defense response to virus was the secondary significant biological pathway, and which included measles, influenza A, hepatitis B virus (HBV), herpes simplex virus (HSV), and Epstein-Barr (EB) virus infection (Fig. 6). The rs34536443 variant may be a bridge linking psoriasis to IPF, rs11795343 is located in the DDX58 gene, which also known as “DExD/H-box helicase 58,” is a gene that encodes a protein called RIG-I (Retinoic acid-inducible gene I), belonging to the DExD/H-box helicase family. RIG-I plays a crucial role in the immune system as a receptor that triggers antiviral immune responses upon viral infection. It plays a pivotal role in safeguarding the body against viral infections and ensuring the homeostasis of the immune system [38]. Disruption or defects in RIG-I function can increase the risk of infections or contribute to the development of autoimmune diseases. DDX58 has been newly identified as a susceptibility gene in psoriasis, it directly causes the production of IL-23 and triggers psoriasis-like skin disease [39]. Certain viruses, such as EBV, CMV, and human herpesvirus (HHV), have been detected in elevated concentrations of individuals with IPF, implicating a potential mechanistic link between latent viral infections and the development of IPF [40, 41]. We assume that DDX58 mediated antiviral immune responses may be one of the potential mechanisms to increase the risk of IPF in psoriasis patients. Furthermore, the rs34536443 variant, located in tyrosine kinase 2 (TYK2) gene, has also been considered to mediate the occurrence of increased risk of IPF. Since TYK2 is a member of the Janus kinase (JAK) family, TYK2 is involved in the intracellular signaling initiated by various cytokines such as IFN, IL-6, IL-12, or IL-23, leading to the phosphorylation of downstream STAT proteins [42, 43]. In psoriasis, the JAK/STAT pathway enhances the inflammatory response by regulating cytokine expression [44]. Likewise, these auto-inflammatory responses may exert a crucial influence on both the initiation and progression of IPF [45].

Therefore, our investigation underscores the potential shared pathways and genetic interplay underlying psoriasis and IPF. These novel genetic insights may have significant implications for understanding the pathogenesis of both diseases and could potentially inform the development of targeted therapies for patients affected by either condition.

However, several limitations to this MR study deserve our attention. First, a measure of disease severity was not available, the proportion of psoriasis cases with different levels of severity will influence the effect size of the outcome. Second, one of the limitations of the UK biobank is the over-estimation of self-reported cases, which can impact the reliability of research conclusions [46]. Indeed, the constraints of coding-based studies encompass inaccuracies in data representation, inherent biases, missing data, and a dearth of clinical details, among other factors. Third, the examined GWASs were primarily conducted in individuals of European ancestry, which might limit the generalization of our findings to other ethnicities, since the incidence of psoriasis varies significantly among different countries. Nonetheless, the European origin reduces the likelihood of population-stratification bias to influence our results. Fourth, genetic factors and environmental factors may have complex interactions, and MR studies may struggle to comprehensively account for these interactions, thus confirming a clear causal association between psoriasis and IPF may require more research.

Conclusion

In conclusion, this MR analysis provided compelling evidence supporting a causally positive association between psoriasis and IPF. Our results underscore the importance of shared genetic factors in the pathogenesis of these two diseases. Further research into the molecular mechanisms connecting psoriasis and IPF may hold the key to developing targeted therapies and improving patient outcomes in both clinical domains.

Data availability

Data available in a publicly accessible repository that does not issue DOIs. Publicly available datasets were analyzed in this study.

References

  1. Parisi R, Iskandar IYK, Kontopantelis E, Augustin M, Griffiths CEM, Ashcroft DM. National, regional, and worldwide epidemiology of psoriasis: systematic analysis and modelling study. BMJ. 2020;369: m1590.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Griffiths CEM, Armstrong AW, Gudjonsson JE, Barker J. Psoriasis. Lancet. 2021;397:1301–15.

    Article  PubMed  CAS  Google Scholar 

  3. Ma F, Plazyo O, Billi AC, Tsoi LC, Xing X, Wasikowski R, Gharaee-Kermani M, Hile G, Jiang Y, Harms PW, et al. Single cell and spatial sequencing define processes by which keratinocytes and fibroblasts amplify inflammatory responses in psoriasis. Nat Commun. 2023;14:3455.

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  4. Fyhrquist N, Muirhead G, Prast-Nielsen S, Jeanmougin M, Olah P, Skoog T, Jules-Clement G, Feld M, Barrientos-Somarribas M, Sinkko H, et al. Microbe-host interplay in atopic dermatitis and psoriasis. Nat Commun. 2019;10:4703.

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  5. Griffiths CE, Barker JN. Pathogenesis and clinical features of psoriasis. Lancet. 2007;370:263–71.

    Article  PubMed  CAS  Google Scholar 

  6. Takeshita J, Grewal S, Langan SM, Mehta NN, Ogdie A, Van Voorhees AS, Gelfand JM. Psoriasis and comorbid diseases: epidemiology. J Am Acad Dermatol. 2017;76:377–90.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Parisi R, Rutter MK, Lunt M, Young HS, Symmons DPM, Griffiths CEM, Ashcroft DM. Psoriasis and the risk of major cardiovascular events: cohort study using the clinical practice research datalink. J Invest Dermatol. 2015;135:2189–97.

    Article  PubMed  CAS  Google Scholar 

  8. Alinaghi F, Tekin HG, Burisch J, Wu JJ, Thyssen JP, Egeberg A. Global prevalence and bidirectional association between psoriasis and inflammatory bowel disease-a systematic review and meta-analysis. J Crohns Colitis. 2020;14:351–60.

    Article  PubMed  Google Scholar 

  9. Li X, Kong L, Li F, Chen C, Xu R, Wang H, Peng S, Zhou M, Li B. Association between psoriasis and chronic obstructive pulmonary disease: a systematic review and meta-analysis. PLoS ONE. 2015;10: e0145221.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Ballardini N. The association between psoriasis and asthma. Br J Dermatol. 2015;172:1485–6.

    Article  PubMed  CAS  Google Scholar 

  11. Wu JJ, Nguyen TU, Poon KY, Herrinton LJ. The association of psoriasis with autoimmune diseases. J Am Acad Dermatol. 2012;67:924–30.

    Article  PubMed  Google Scholar 

  12. Makredes M, Robinson D Jr, Bala M, Kimball AB. The burden of autoimmune disease: a comparison of prevalence ratios in patients with psoriatic arthritis and psoriasis. J Am Acad Dermatol. 2009;61:405–10.

    Article  PubMed  Google Scholar 

  13. Ishikawa G, Dua S, Mathur A, Acquah SO, Salvatore M, Beasley MB, Padilla ML. Concomitant interstitial lung disease with psoriasis. Can Respir J. 2019;2019:5919304.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Butt YM, Smith ML, Tazelaar HD, Roden AC, Mengoli MC, Larsen BT. Surgical pathology of diffuse parenchymal lung disease in patients with psoriasis or psoriatic arthritis. Arch Pathol Lab Med. 2022;147:525–33.

    Article  PubMed  Google Scholar 

  15. Okubo Y, Kotowsky N, Gao R, Saito K, Morita A. Clinical characteristics and health-care resource utilization in patients with generalized pustular psoriasis using real-world evidence from the Japanese Medical Data Center database. J Dermatol. 2021;48:1675–87.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Spagnolo P, Kropski JA, Jones MG, Lee JS, Rossi G, Karampitsakos T, Maher TM, Tzouvelekis A, Ryerson CJ. Idiopathic pulmonary fibrosis: disease mechanisms and drug development. Pharmacol Ther. 2021;222: 107798.

    Article  PubMed  CAS  Google Scholar 

  17. Richeldi L, Collard HR, Jones MG. Idiopathic pulmonary fibrosis. Lancet. 2017;389:1941–52.

    Article  PubMed  Google Scholar 

  18. O’Dwyer DN, Ashley SL, Gurczynski SJ, Xia M, Wilke C, Falkowski NR, Norman KC, Arnold KB, Huffnagle GB, Salisbury ML, et al. Lung microbiota contribute to pulmonary inflammation and disease progression in pulmonary fibrosis. Am J Respir Crit Care Med. 2019;199:1127–38.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Mleczko M, Gerkowicz A, Krasowska D. Chronic inflammation as the underlying mechanism of the development of lung diseases in psoriasis: a systematic review. Int J Mol Sci. 2022;23:1767.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Zhang X, Shi L, Sun T, Guo K, Geng S. Dysbiosis of gut microbiota and its correlation with dysregulation of cytokines in psoriasis patients. BMC Microbiol. 2021;21:78.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Gupta R, Espiritu J. Azathioprine for the rare case of nonspecific interstitial pneumonitis in a patient with psoriasis. Ann Am Thorac Soc. 2015;12:1248–51.

    PubMed  Google Scholar 

  22. Matsumoto Y, Abe N, Tobita R, Kawakami H, Nakayama H, Setoguchi Y, Tsuboi R, Okubo Y. The risk of interstitial lung disease during biological treatment in Japanese patients with psoriasis. Clin Exp Dermatol. 2020;45:853–8.

    Article  PubMed  CAS  Google Scholar 

  23. Emdin CA, Khera AV, Kathiresan S. Mendelian randomization. JAMA. 2017;318:1925–6.

    Article  PubMed  Google Scholar 

  24. Davies NM, Holmes MV, Davey Smith G. Reading Mendelian randomisation studies: a guide, glossary, and checklist for clinicians. BMJ. 2018;362: k601.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Bowden J, Del Greco MF, Minelli C, Zhao Q, Lawlor DA, Sheehan NA, Thompson J, Davey Smith G. Improving the accuracy of two-sample summary-data Mendelian randomization: moving beyond the NOME assumption. Int J Epidemiol. 2018;48:728–42.

    Article  PubMed Central  Google Scholar 

  26. Burgess S, Bowden J, Fall T, Ingelsson E, Thompson SG. Sensitivity analyses for robust causal inference from Mendelian randomization analyses with multiple genetic variants. Epidemiology. 2017;28:30–42.

    Article  PubMed  Google Scholar 

  27. Bowden J, Davey Smith G, Burgess S. Mendelian randomization with invalid instruments: effect estimation and bias detection through Egger regression. Int J Epidemiol. 2015;44:512–25.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Kawamoto H, Hara H, Minagawa S, Numata T, Araya J, Kaneko Y, Umezawa Y, Asahina A, Nakagawa H, Kuwano K. Interstitial pneumonia in psoriasis. Mayo Clin Proc Innov Qual Outcomes. 2018;2:370–7.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Kharibam P, Laishram S, Choudhary N, Choudhury A, Kant R. Concomitant interstitial lung disease with psoriasis. Cureus. 2022;14: e26979.

    PubMed  PubMed Central  Google Scholar 

  30. Nie YJ, Wu SH, Xuan YH, Yan G. Role of IL-17 family cytokines in the progression of IPF from inflammation to fibrosis. Mil Med Res. 2022;9:21.

    PubMed  PubMed Central  CAS  Google Scholar 

  31. Fraser E, Denney L, Antanaviciute A, Blirando K, Vuppusetty C, Zheng Y, Repapi E, Iotchkova V, Taylor S, Ashley N, et al. Multi-modal characterization of monocytes in idiopathic pulmonary fibrosis reveals a primed type I interferon immune phenotype. Front Immunol. 2021;12: 623430.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Latsi P, Pantelidis P, Vassilakis D, Sato H, Welsh KI, du Bois RM. Analysis of IL-12 p40 subunit gene and IFN-gamma G5644A polymorphisms in Idiopathic Pulmonary Fibrosis. Respir Res. 2003;4:6.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Trinchieri G, Gerosa F. Immunoregulation by interleukin-12. J Leukoc Biol. 1996;59:505–11.

    Article  PubMed  CAS  Google Scholar 

  34. Tsoutsou PG, Gourgoulianis KI, Petinaki E, Germenis A, Tsoutsou AG, Mpaka M, Efremidou S, Molyvdas PA. Cytokine levels in the sera of patients with idiopathic pulmonary fibrosis. Respir Med. 2006;100:938–45.

    Article  PubMed  Google Scholar 

  35. Otaki N, Motomura Y, Terooatea T, Thomas Kelly S, Mochizuki M, Takeno N, Koyasu S, Tamamitsu M, Sugihara F, Kikuta J, et al. Activation of ILC2s through constitutive IFNγ signaling reduction leads to spontaneous pulmonary fibrosis. Nat Commun. 2023;14:8120.

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  36. Das NA, Carpenter AJ, Yoshida T, Kumar SA, Gautam S, Mostany R, Izadpanah R, Kumar A, Mummidi S, Siebenlist U, Chandrasekar B. TRAF3IP2 mediates TWEAK/TWEAKR-induced pro-fibrotic responses in cultured cardiac fibroblasts and the heart. J Mol Cell Cardiol. 2018;121:107–23.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Wang W, Bale S, Wei J, Yalavarthi B, Bhattacharyya D, Yan JJ, Abdala-Valencia H, Xu D, Sun H, Marangoni RG, et al. Fibroblast A20 governs fibrosis susceptibility and its repression by DREAM promotes fibrosis in multiple organs. Nat Commun. 2022;13:6358.

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  38. Onomoto K, Onoguchi K, Yoneyama M. Regulation of RIG-I-like receptor-mediated signaling: interaction between host and viral factors. Cell Mol Immunol. 2021;18:539–55.

    Article  PubMed  CAS  Google Scholar 

  39. Zhu H, Lou F, Yin Q, Gao Y, Sun Y, Bai J, Xu Z, Liu Z, Cai W, Ke F, et al. RIG-I antiviral signaling drives interleukin-23 production and psoriasis-like skin disease. EMBO Mol Med. 2017;9:589–604.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Sheng G, Chen P, Wei Y, Yue H, Chu J, Zhao J, Wang Y, Zhang W, Zhang HL. Viral infection increases the risk of idiopathic pulmonary fibrosis: a meta-analysis. Chest. 2020;157:1175–87.

    Article  PubMed  CAS  Google Scholar 

  41. Yonemaru M, Kasuga I, Kusumoto H, Kunisawa A, Kiyokawa H, Kuwabara S, Ichinose Y, Toyama K. Elevation of antibodies to cytomegalovirus and other herpes viruses in pulmonary fibrosis. Eur Respir J. 1997;10:2040–5.

    Article  PubMed  CAS  Google Scholar 

  42. Wrobleski ST, Moslin R, Lin S, Zhang Y, Spergel S, Kempson J, Tokarski JS, Strnad J, Zupa-Fernandez A, Cheng L, et al. Highly selective inhibition of tyrosine kinase 2 (TYK2) for the treatment of autoimmune diseases: discovery of the allosteric inhibitor BMS-986165. J Med Chem. 2019;62:8973–95.

    Article  PubMed  CAS  Google Scholar 

  43. Schwartz DM, Bonelli M, Gadina M, O’Shea JJ. Type I/II cytokines, JAKs, and new strategies for treating autoimmune diseases. Nat Rev Rheumatol. 2016;12:25–36.

    Article  PubMed  CAS  Google Scholar 

  44. Nogueira M, Puig L, Torres T. JAK inhibitors for treatment of psoriasis: focus on selective TYK2 inhibitors. Drugs. 2020;80:341–52.

    Article  PubMed  CAS  Google Scholar 

  45. Yuan S, Wang L, Zhang H, Xu F, Zhou X, Yu L, Sun J, Chen J, Ying H, Xu X, et al. Mendelian randomization and clinical trial evidence supports TYK2 inhibition as a therapeutic target for autoimmune diseases. EBioMedicine. 2023;89: 104488.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Leavy OC, Allen RJ, Kraven LM, Morgan AD, Tobin MD, Quint JK, Jenkins RG, Wain LV. The use of genetic information to define idiopathic pulmonary fibrosis in UK biobank. Chest. 2023;163:362–5.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We would like to thank the authors and participants of all GWAS from which we used summary statistics data.

Funding

This study was supported by the National Natural Science Foundation of China (No.81873851 and No.82170019).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, L-LC and Y-JW; methodology, M-JH and L-LC; software, Y-JW; validation, L-LC and X-RZ; formal analysis, M-JH, Y-LL and Y-JW; investigation, M-JH, Y-JW,L-LC, and X-RZ; resources, X-RZ; data curation, Y-JW and Y-LL; writing—original draft preparation, L-LC; writing—review and editing, M-JH, Y-JW, Y-LL and X-RZ; visualization, M-JH; supervision, Y-JW; project administration, Y-JW; funding acquisition, X-RZ. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Xiangrong Zheng.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

All authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, L., Wei, Y., Hu, M. et al. Psoriasis may increase the risk of idiopathic pulmonary fibrosis: a two-sample Mendelian randomization study. Respir Res 25, 101 (2024). https://doi.org/10.1186/s12931-024-02721-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12931-024-02721-5

Keywords