Next Article in Journal
Is Cadmium Toxicity Tissue-Specific? Toxicogenomics Studies Reveal Common and Specific Pathways in Pulmonary, Hepatic, and Neuronal Cell Models
Next Article in Special Issue
Profiling the Atopic Dermatitis Epidermal Transcriptome by Tape Stripping and BRB-seq
Previous Article in Journal
The Role of AGE-RAGE Signalling as a Modulator of Gut Permeability in Diabetes
Previous Article in Special Issue
Neutrophil Pathways of Inflammation Characterize the Blood Transcriptomic Signature of Patients with Psoriasis and Cardiovascular Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Chronic Inflammation as the Underlying Mechanism of the Development of Lung Diseases in Psoriasis: A Systematic Review

Department of Dermatology, Venerology and Pediatric Dermatology, Medical University of Lublin, 20-081 Lublin, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(3), 1767; https://doi.org/10.3390/ijms23031767
Submission received: 11 January 2022 / Revised: 30 January 2022 / Accepted: 2 February 2022 / Published: 4 February 2022
(This article belongs to the Special Issue Molecular Research on Inflammatory Skin Diseases)

Abstract

:
Psoriasis is a systemic inflammatory disease caused by dysfunctional interactions between the innate and adaptive immune responses. The systemic inflammation in psoriasis may be associated with the development of comorbidities, including lung diseases. In this review, we aimed to provide a summary of the evidence regarding the prevalence of lung diseases in patients with psoriasis and the potential underlying mechanisms. Twenty-three articles published between March 2010 and June 2021 were selected from 195 initially identified records. The findings are discussed in terms of the prevalence of asthma, chronic obstructive pulmonary disease, interstitial lung disease, obstructive sleep apnea, pulmonary hypertension, and sarcoidosis in psoriasis. A higher prevalence of lung diseases in psoriasis has been confirmed in asthma, chronic obstructive pulmonary disease, obstructive sleep apnea, and pulmonary hypertension. These conditions are important as they are previously unrecognized causes of morbidity and mortality in psoriasis. The development of lung diseases in patients with psoriasis can be explained by several mechanisms, including common risk factors, shared immune and molecular characteristics associated with chronic inflammation, as well as other mechanisms. Understanding the prevalence of lung diseases in psoriasis and their underlying mechanisms can help implement appropriate preventative and therapeutic strategies to address respiratory diseases in patients with psoriasis.

1. Introduction

Psoriasis is a systemic inflammatory disease affecting 2–4% of the population that develops as a result of abnormal interactions between the innate and adaptive immune responses [1]. The classical manifestations of psoriasis include raised areas of inflamed skin covered with silvery-white scales, mainly over the extensor surfaces of the limbs, on the scalp, and within the lumbosacral region [2]. The disease can also affect the joints, causing psoriatic arthritis [3].
The systemic inflammation in psoriasis may be associated with the development of comorbidities [4]. Interestingly, concomitant conditions can already be detectable in children and adolescents with the disease [5]. Patients with psoriasis have an increased risk of hyperlipidemia [6], hypertension [7], diabetes [8], and an increased body mass index (BMI) [9]. Furthermore, psoriasis has been associated with a higher prevalence of gastrointestinal [10] disorders and chronic kidney disease [11].
Recent studies also suggest an increased incidence of lung diseases in psoriasis. The development of lung diseases in populations with psoriasis may be due to several mechanisms, including exposure to common risk factors, increased susceptibility to respiratory diseases in psoriasis, or other mechanisms (Figure 1).
Lung diseases adversely affect the quality of life of patients with psoriasis and contribute to the overall burden of disease. In this review, we aimed to provide a summary of the current evidence regarding the prevalence of lung diseases in patients with psoriasis and the potential underlying mechanisms.

2. Materials and Methods

We carried out a comprehensive review of PubMed- and EMBASE (Elsevier, Amsterdam, The Netherlands)-indexed articles in order to assess the prevalence of lung diseases in psoriasis, as well as to identify factors that could explain the seemingly higher prevalence of lung diseases in patients with psoriasis. This systematic review was conducted in accordance with the preferred reporting items for systematic reviews and meta-analyses (PRISMA) statement.

2.1. Search Strategy

A comprehensive review of PubMed- and EMBASE-indexed articles was carried out. Our literature search strategy included selected search terms combined using the following Boolean operators of “AND” and “OR”: (asthma) OR (chronic obstructive pulmonary disease) OR (obstructive sleep apnea) OR (pulmonary hypertension) OR (interstitial lung disease) OR (sarcoidosis) AND (psoriasis). Search terms were identified using the MeSH thesaurus. To minimize any potential bias, there were no limits on the language of publication.

2.2. Selection Criteria

The authors developed a list of studies during the selection process. The search was narrowed by adding a filter for publication dates between March 2010 and September 2021. We also obtained additional studies from the reference lists of relevant reviews. Subsequently, the references and abstracts of articles found on PubMed and EMBASE were screened to identify potentially relevant publications. Next, the full texts of the identified articles were retrieved, and the articles were reviewed to narrow the selection to relevant reports. This stage served as an in-depth verification of findings. Eligible studies included the following: clinical trials, observational studies, meta-analyses, systematic reviews, case series, and case reports published during the predefined period. The exclusion criteria included the following: serious weaknesses in study design, e.g., failure to choose a control group or the use of an inappropriate control group in studies that weren’t case reports/case series; articles where the full text was not retrievable; and articles where the outcomes could not be attributed to device.
Once the selection of the articles was completed and all the full texts of the selected articles were retrieved, data extraction followed. The methodology used in the literature search and review is detailed in Figure 2 and Figure 3.

2.3. Data Extraction

A total of 23 relevant publications published from March 2010–September 2021 were selected during the PubMed- and EMBASE-indexed literature search. The authors extracted data from the included studies using a standard form. Data on the following were extracted: (1) basic characteristics of the included studies (full reference, publication year, country, ethnicity, number of patients, age range, percentage of male patients, and lung disease); (2) prevalence rates of lung diseases in question. The domains would be rated for their appropriateness as yes, partly, unsure, or no, respectively.

3. Results

A total of 195 references were identified through electronic database searches and searched by title and abstract. Following the exclusion of duplicate and irrelevant references, a total of 128 articles were considered essential, reviewed in their full text, and selected or rejected based on the exclusion criteria. All articles were reviewed according to the scheme shown in Figure 2. A total of 23 original reports that met the inclusion and exclusion criteria were analyzed, including one case report, one case series, seventeen retrospective reviews, zero prospective studies, zero comparative studies, and four RCTs. Table 1 summarizes the studies included in the analysis. Of the 195 articles included in the review, 43 related to asthma, 18 to chronic obstructive pulmonary disease (COPD), 26 to obstructive sleep apnea (OSA), 3 to pulmonary atrial hypertension (PAH), 53 to interstitial lung disease (ILD), and 52 to sarcoidosis.

4. Discussion

4.1. Prevalence of Lung Diseases in Psoriasis

A number of studies reported an increased prevalence of asthma [12], chronic obstructive pulmonary disease [20,21], obstructive sleep apnea [25], and pulmonary hypertension [31] in psoriasis patients. There are also single studies that report an increased prevalence of interstitial lung disease and sarcoidosis in psoriasis [32,34]. To our knowledge, ours is the first systematic review to discuss the prevalence and pathophysiology of lung diseases in psoriasis.
The risk of developing individual lung diseases in psoriasis varies with sex, age, and the severity of psoriasis. In terms of sex, a Taiwanese trusted source study found an increased risk of COPD in men with psoriasis (HR = 2.38, 95% CI: 1.36–4.18) [23]. There were no similar relationships found for other lung diseases. Older patients with psoriasis (≥50 years) had a higher risk of asthma (OR = 1.64, 95% CI: 1.44–1.88) and chronic obstructive pulmonary disease (HR = 2.19, 95% CI: 1.27–3.77) than younger ones [12,23]. In children, observational studies suggest an increased risk of asthma compared to the general population, but the data are inconsistent [13,17,18,19]. Furthermore, the increased susceptibility to asthma in children was associated with moderate to severe psoriasis [17].
In terms of psoriasis severity, it was shown that even those with mild psoriasis had an increased risk of developing COPD (OR = 1.66, 95% CI: 1.00–2.76) [21] and sarcoidosis (HR = 1.49, 95% CI: 1.18–1.87); however, moderate to severe psoriasis also showed an increased risk of developing COPD (OR = 2.15, 95% CI: 1.26–3.67) [21], pulmonary hypertension (OR = 1.46, 95% CI: 1.09–1.94) [31], and sarcoidosis (HR = 2.51, 95% CI: 1.64–3.85) [34]. An increased risk of pulmonary hypertension has not been observed in patients with mild psoriasis [31]. There was no association between psoriasis severity and an increased risk of asthma [12] or OSA [35].
Certain lung diseases, such as interstitial lung disease [32] and sarcoidosis [34], have an uncertain relationship with psoriasis. Interstitial lung disease was occasionally reported in patients with psoriasis, with most cases identified as immunosuppressant-induced pneumonitis [36,37,38,39,40,41]. In a study on patients with interstitial lung disease not treated with immunosuppressants, 4.7% were also diagnosed with psoriasis [32]. However, another study showed that pulmonary fibrosis was not more common in psoriasis than would be expected in the general population [42]. In one study, researchers found that people with psoriasis were more predisposed to developing sarcoidosis than controls [34]. Further large prospective studies are warranted to investigate the association between psoriasis and both interstitial lung disease as well as sarcoidosis.
The main issue with studies that assess the association between psoriasis and lung diseases seems to be their disregard of common risk factors associated with both respiratory problems and psoriasis. Failure to adjust for these factors may result in risk overestimation of the discussed lung diseases. Thus, additional well-designed studies are urgently needed to address the potential contribution of pulmonary risk factors to the development of lung diseases in patients with psoriasis.

4.2. Common Risk Factors

Common risk factors, responsible for both the development and worsening of lung diseases and psoriasis, include smoking, obesity and low physical activity, pollutants, infections, exposure to allergens, malnutrition, metabolic syndrome, connective tissue disorders, depression, and the use of certain medications (Figure 4).

4.2.1. Smoking

Research shows that patients with psoriasis report smoking, both passive and active, more often than the general population [63]. Smoking has been shown to significantly correlate with both the development and severity of psoriatic lesions [64]. At the same time, smoking is recognized as the most important causative factor in chronic obstructive pulmonary disease [43]. Other lung diseases that have been linked to smoking include asthma [44] and idiopathic pulmonary fibrosis [45]. Thus, smoking can be a factor that explains the increased prevalence of lung disease in patients with psoriasis.

4.2.2. Obesity and Low Physical Activity

Obesity and low physical activity have been identified as risk factors for the development and severity of psoriasis, and a higher prevalence of obesity has been shown in patients with psoriasis [65,66]. Obesity is a principal risk factor for the development of respiratory diseases such as asthma [55], pulmonary hypertension, and sleep apnea [56]. It has also been implicated in chronic obstructive pulmonary disease [57]. Thus, obesity may explain the higher prevalence of lung disease in psoriasis.

4.2.3. Pollutants

Cadmium, the element found in tooth fillings, used in battery and television manufacturing, as well as the aircraft industry, has been proposed as an air pollutant linked to the development of psoriasis. Higher levels of cadmium in the blood have been observed in patients with psoriasis [67]. At the same time, a higher blood cadmium level was associated with chronic obstructive pulmonary disease in males, including those who had never smoked [62]. Thus, cadmium (and potentially other air pollutants) can equally impact the development of psoriasis and lung diseases in the same individuals.

4.2.4. Infections

Infections can play a role in both psoriasis and lung diseases. The altered microbiome in the lower respiratory tract, which can increase the risk of psoriasis, interacts with the innate mucosal immune system and has been implicated in the development of asthma [18,60]. Furthermore, human immunodeficiency virus (HIV) is a well-known risk factor associated with psoriasis and lung diseases such as asthma, chronic obstructive pulmonary disease, and pulmonary arterial hypertension [59,61,68]. It is possible that HIV infection contributes to the development or worsening of both psoriasis and lung diseases.

4.2.5. Allergy

Patients with psoriasis have been shown to present weakly expressed hypersensitivity to some inhalant, food, and contact allergens, such as birch, artemisia, timothy grass and rye pollens, house dust mites, and molds [69]. The intensity of hypersensitivity reactions correlated with PASI [69]. Sensitization to allergens also plays an important role in the development, severity, and treatment of asthma. Recent studies have found that an allergy to house dust mites, pet dander, cockroaches, or fungi is a risk factor for asthma [58].

4.2.6. Malnutrition

The composition of the intestinal microbiota can influence the susceptibility to an increased incidence of many inflammatory diseases. Body mass index and the consumption of red wine were shown to affect the intestinal microbial composition. Diet-induced dysbiosis of intestinal microbiota may induce cytokine imbalance, which is the underlying mechanism in psoriasis [70]. However, dysbiosis of the intestinal microbiota has also been implicated in asthma through early life antimicrobial exposure, Caesarian birth, formula feeding, lack of maternal exposure to pets or livestock during pregnancy, and maternal consumption of antimicrobials during pregnancy [51].

4.2.7. Metabolic Syndrome and Its Components

A higher prevalence of hyperlipidemia [6], hypertension [7], and diabetes mellitus [8] was shown in patients with psoriasis. Hyperlipidemia is also associated with poorer hyperinflation and airway obstruction in patients with chronic obstructive pulmonary disease [52]. Obstructive sleep apnea is common in patients with hypertension (a prevalence rate of 37% to 56%), and the prevalence rate in those with resistant hypertension can be as high as 70% to 83% [53]. Individuals with diabetes are at an increased risk of the following several pulmonary conditions: asthma, COPD, PAH, and fibrosis [54]. Thus, the individual components of metabolic syndrome may explain the increased risk of lung disease in psoriasis.

4.2.8. Connective Tissue Disorder

It has been accepted that patients with an autoimmune disease have a higher risk of developing another autoimmune disorder [71]. Whereas the data on the coexistence of psoriasis and systemic lupus erythematous are unclear [72], systemic sclerosis was found to be independently associated with psoriasis [73]. Furthermore, the leading causes of death in systemic sclerosis are pulmonary arterial hypertension and interstitial lung diseases [49,50]. Hence, connective tissue disorder, which shares an autoimmune etiology with psoriasis, may lead to lung disease in its late stages, thus explaining the link between psoriasis and lung disease.

4.2.9. Depression

Psoriasis is a chronic inflammatory dermatosis that limits patients’ ability to work and socialize, affecting their family life as well as leisure and sexual activity [74]. These limitations can be a source of stress, which is a well-established trigger of psoriasis [75]. On the other hand, depression is a common and chronic comorbidity in patients with interstitial lung disease [48]. The use of antidepressants has been associated with a significantly increased risk of idiopathic pulmonary arterial hypertension [46]. Moreover, in people suffering from depression, previous studies have shown an increase in proinflammatory cytokines such as TNFα and IL-6, similar to that in psoriasis [76]. In this way, depression can be a link between psoriasis (which leads to depression) and lung disease (which may, for instance, develop in response to the treatment of depression with selective serotonin reuptake inhibitors).

4.2.10. Medications

Apart from the previously mentioned SSRIs, which have been linked to idiopathic pulmonary arterial hypertension [46], there are medications that can induce bronchospasm, e.g., β-blockers, angiotensin-converting enzyme (ACE) inhibitors, nonsteroidal anti-inflammatory drugs (NSAIDs), and interferons [47]. Interestingly, many of them, that is, β-blockers, angiotensin-converting enzyme inhibitors, nonsteroidal anti-inflammatory drugs, lithium, anti-malarial drugs, interferons, imiquimod, terbinafine, tetracycline, and fibrate drugs [77], have been mentioned in the context of drug-related psoriasis, referring to the onset or exacerbation of psoriasis associated with certain medications. Thus, medications can contribute to the development or exacerbation of both psoriasis and certain lung diseases in patients prescribed the above treatments for other indications.

4.2.11. Other Factors

There was a single report of erythrodermic pustular psoriasis induced by intravesical bacillus Calmette-Guérin immunotherapy in a patient with bladder cancer [78]. There was also a single report of granulomatous pneumonia as a complication of bacillus Calmette-Guérin immunotherapy [79]. These show that there may also be other common factors, poorly understood to date, that lung diseases and psoriasis have in common. Thus, further research is warranted.

4.3. Chronic Inflammation as the Underlying Mechanism of the Development of Lung Diseases in Psoriasis

Systemic inflammation in psoriasis triggers immune changes that can lead to the development of comorbidities [80], and patients with an inflammatory autoimmune disease have a higher risk of developing another autoimmune disorder [71,81]. So far, no studies have been conducted to evaluate the role of immunological mechanisms and inflammatory mediators in the pathogenesis of lung disease in patients with psoriasis. However, elevated levels of many pro-inflammatory cytokines have been found in skin lesions as well as in blood, but only in moderate to severe psoriasis, as measured by the PASI (Psoriasis Area and Severity Index) and defined as more than 42% of the body surface area [82]. It is therefore possible that, due to its possible autoimmune component, psoriasis-related immune dysfunction could trigger an abnormal immune response in the lungs, acting de novo on the lung tissue and exacerbating the pre-existing inflammation caused by the underlying disease [83,84,85,86]. Tumor necrosis factor-α, dendritic cells, interleukin-1, -6, and -8, and T-cells all contribute substantially to the pathogenesis of psoriasis and increase the risk of other systemic diseases [87]. The increased levels of inflammatory markers, such as tumor necrosis factor-α (TNF-α), interleukin-6, and C-reactive protein, have indeed been found in the lung tissue of patients with psoriasis [88].
Lung diseases share many aspects with psoriasis (Figure 5). The similarities include immune processes, cytokine profiles, and immune cell types. The immune profile of psoriasis is mediated by T helper 1 (Th1) and Th17 cells, but also by Th22 and Th9 activation [89]. Interestingly, neutrophilic inflammation in severe or corticosteroid-resistant asthma is mediated by Th1 and Th17 cells, as well as other neutrophilic mediators [90]. The same cells accumulate in the lungs of patients with stable chronic obstructive pulmonary disease [91,92] or in active disease sites, forming non-keratinizing granulomas in patients with sarcoidosis [93]. Th17 cells were also implicated in one of the most common pathways leading to alveolitis in interstitial lung disease [94].

4.3.1. Th1 Cells

Th1 cell activation leads to an increase in interferon γ (INF-γ) and tumor necrosis factor α (TNF-α) levels [95]. INF-γ- and TNF-α-mediated inflammatory signaling is the key process in psoriasis [96]. However, elevated levels of the same inflammatory cytokines have also been found in the sputum and bronchopulmonary lavage fluid of patients with chronic obstructive pulmonary disease [97,98], as well as in the serum of patients with obstructive sleep apnea [97,98,99,100]. There are also data to support the role of INF-γ and TNF-α in pulmonary emphysema [101,102]. Moreover, TNF-α may upregulate goblet cell hyperplasia, which has been implicated in a number of lung diseases [103]. The above evidence shows that Th1 cells, as common mediators of inflammation in both psoriasis and lung diseases, add to the explanation of their concomitance.

4.3.2. Th17 Cells

Th17 cells produce IL-17A, -17F, and -22, which are all known as highly inflammatory cytokines that induce the keratinocyte activation and proliferation seen in psoriasis [76,85,86,104]. The increased activity of the same cytokines has been observed in the sputum of patients with asthma [105], in bronchoalveolar lavage fluid of patients with sarcoidosis [106], as well as in the serum of patients with obstructive sleep apnea [107,108,109] and sarcoidosis [106]. The IL-17-high asthma phenotype, with bronchial epithelial dysfunction and an upregulated inflammatory response, resembles that of psoriasis [110]. In the murine lung epithelium, IL-17 overexpression led to inflammation with a chronic obstructive pulmonary disease-like phenotype involving CD4 cell recruitment, mucus hypersecretion, small airway fibrosis, and chemokine expression [94,111,112]. Thus, Th17 cell involvement could add to the understanding of links between psoriasis and lung disease.

4.3.3. Th22 Cells

Th22 cells are identified by the production of IL-22, which is one of the members of the IL-10 family [113]. IL-22 has been implicated in both psoriasis and several lung diseases. In psoriasis, it is known to promote keratinocyte activation and the formation of epidermal acanthosis [114,115]. Elevated levels of IL-22 were found in patients with T-cell-mediated lung diseases, such as interstitial lung disease [116]. IL-22 has also been shown to induce smoking-dependent airway remodeling and emphysema-like alveolar enlargement in chronic obstructive pulmonary disease [117], as well as the recruitment, activation, and proliferation of mononuclear cells, contributing to alveolitis and granuloma formation in sarcoidosis [118].

4.3.4. Th9 Cells

Th9 cells are the most recently defined subset of T-helper cells. They produce IL-9, which might be involved in immune-mediated disease [119]. The exact role of Th9 cells in the pathogenesis of psoriasis and lung diseases is still poorly understood, which warrants further research. It is postulated that Th9 cells may participate in initiating and maintaining skin inflammation [120]. IL-9 overexpression in the lungs of transgenic mice appears to enhance airway inflammation and play a role in the early initiation of airway remodeling, most notably mucus production within the epithelium [121]. Thus, alongside other cell subsets, Th9 cells can help explain the link between psoriasis and lung diseases.

4.3.5. Other Immune Mechanisms

Other immune mechanisms implicated in the pathogenesis of psoriasis and lung diseases include transcription factors (NF-κB, HIF-1α) [104]; T-cell receptor and co-stimulatory molecules (e.g., CD3, CD8) [84,122]; T-cell cytokines (e.g., interferon-γ, I L-13, IL-17, and IL-23) [83,84] and other inflammatory cytokines (e.g., TGF-β, IL-12, IL-18, IL-22, VEGF, and TRAIL) [84,123,124,125,126,127,128], chemokines and receptors (e.g., CXCR1, CXCR2, and CXCR3) [84,129], adhesion molecules (e.g., ICAM-1, VCAM-1, E-selectin, and CD18) [84,127,128,130], and proteases (elastase, cathepsins, and matrix metalloproteinases) [84,131]. They are believed to contribute to pathological changes in the lungs of patients with psoriasis, which are shown in Figure 6.

4.4. Other Postulated Causal Mechanisms

4.4.1. Genetic Factors and Genetic Instability

A potential genetic link between psoriasis and asthma, chronic obstructive pulmonary disease, or sarcoidosis has been proven. Single-nucleotide polymorphism (SNP)-based genome-wide association studies (GWASs) have identified common variants determining susceptibility to psoriasis and asthma [132,133,134]. In a pathway analysis, genes that were differentially expressed in asthma patients with high levels of IL-17 were shared with genes reported as being altered in psoriasis [110]. Another confirmation of the shared genetic background between psoriasis and asthma is a study conducted on twins, which, using a bivariate variance components twin model, demonstrated an increased occurrence of asthma in patients with psoriasis [16]. Genetic analyses revealed a copy number variation of the β-defensin genes in both psoriasis [135] and chronic obstructive pulmonary disease [136]. Furthermore, the polymorphism in the promoter region of the TNF-alpha gene [137], previously established in psoriasis, was also found in patients with chronic obstructive pulmonary disease [138]. Sarcoidosis, alongside several other autoimmune disorders, including psoriasis, has been demonstrated to be associated with IL23R polymorphism [139,140]. All these findings support the genetic links between psoriasis and lung diseases.
When analyzing the common genetic pathways of psoriasis and lung diseases, the phenomenon of genetic instability should be mentioned, which can be observed as cytogenetic instability and/or molecular instability.
Cytogenetic instability mainly relates to chromosomal instability (CNI), with a high number of breaks and/or numerical and structural chromosome aberrations. DNA damage, hereinafter referred to as the genetic DNA fragment between sister chromatids, can be detected using the analysis of sister chromatid exchange (SCE) [141]. In one study, significantly higher SCE rates were observed in the peripheral lymphocytes of patients with psoriasis [142]. Variations in SCE have also been found in other chronic diseases, but COPD does not appear to be associated with DNA damage [143].
Molecular instability is associated with microsatellite instability (MSI)—as an alteration of allele length due to a change in the number of nucleotide repeats, and/or allelic instability, as a loss of heterozygosity (LOH)—the loss of one of two examined alleles. In addition, MSI and LOH have been reported in a number of human malignancies [141]. Additionally, MSI and LOH have been detected in asthma, COPD, sarcoidosis, and IPF [144,145,146,147]. In part of these studies, specific regions of chromosomes 3p, 5q, 6p, 13q, 14q, and 17q containing (a) genes for susceptibility to asthma and allergy, as well as (b) genes associated with an increased risk of developing irreversible airway obstruction were analyzed [145]. There are no studies on the occurrence of MSI in psoriasis. One study found that specific LOH-altered genetic loci on chromosomes 3p, 7p/q, and 8p are important in the development of psoriatic plaques, but recent research indicates that psoriasis is not related to LOH [148,149]. Based on these results, it has been hypothesized that cigarette-smoke-induced acquired somatic mutations are a major contributor to COPD. Another possible hypothesis may be that MSI reflects a defect in the DNA repair process caused by the oxidative stress associated with smoking, which leads to insufficient airway remodeling [150]. Smoking is believed to cause potentially irreversible genetic changes in epithelial cells [151]. It is possible that cigarette smoking, which exacerbates psoriasis, may cause similar changes in genetic instability.

4.4.2. Epigenetic Mechanisms

Epigenetic processes represent potential molecular routes between genetic backgrounds and environmental risk factors contributing to the pathogenesis of psoriasis and pulmonary diseases. Recent studies highlight the role of epigenetic processes in inflammatory diseases, including psoriasis [152]. Similarly, evidence of aberrant epigenetic marks has emerged in patients with asthma, chronic obstructive pulmonary disease (COPD), and pulmonary arterial hypertension (PAH). Epigenetic mechanisms include the regulation of gene expression at the transcriptional (via DNA methylation and histone modification) and post-transcriptional levels (via long non-coding RNAs and microRNAs) [153].
There are numerous reports on the role of histone modification in psoriasis. Psoriasis is associated with the incorrect expression of histone acetyltransferases (HATs) and histone deacetylases (HDACs), which regulate the balance between histone acetylation and deacetylation. In patients with psoriasis, elevated mRNA levels of HDAC1 compared to healthy controls were noted [154]. Aberrant HDAC activities were also observed in pulmonary fibrosis [155]. Moreover, Ding et al. noted that the expression of HDAC1 and HDAC2 was upregulated in mice exposed to cigarette smoke, a common risk factor for psoriasis and lung diseases [156]. Other studies concerned the mechanisms of histone modulation in controlling cytokine production and its role in the onset of psoriasis and asthma. It has been found that among psoriasis patients, H3K4 methylation is increased in peripheral blood mononuclear cells [157]. The genome-wide mapping of histone modifications in CD4 memory as well as TH1 and TH2 cells revealed differential enrichment of dimethylated lysine 4 of histone H3 (H3K4me2) in TH2 enhancers associated with susceptibility to asthma [158]. It has also been noted that increased mRNA levels of DEFB1 (beta-defensin 1) are correlated with H3K4me3 in the progression of COPD [159].
MicroRNA (miRNA) plays an important role in the pathogenesis of psoriasis. In psoriasis-affected keratinocytes, the increased expression of miR-146a, miR-203, miR-21, miR-31, miR-184, miR-221, and miR-222 was observed, whereas the expression of miR-99a, miR-424, and miR-125b was decreased [160]. MiR-146a, along with miR-146b, participates in homeostasis and controls inflammatory responses in the skin [161]. Moreover, they are negative regulators of inflammatory gene expression in lung tissue [162]. The consistent upregulation of miR-146b was observed in a murine model study of acute and chronic asthma [163]. In the skin, the increase in miR-203 is consistent with the downregulation of suppressor of cytokine signaling-3 (SOCS-3) and the subsequent upregulation of signal transducer and activator of transcription-3 (STAT-3) [164,165]. The transcriptional downregulation of SOCS3 has been observed in asthma and COPD patients [166,167]. The expression of miR-21 is increased in skin affected by psoriasis due to T-cell infiltration [168]. MiR-21 has been associated with the activation of fibroblasts and their differentiation to myofibroblasts in the pathogenesis of asthma, PAH, and IPF [169,170,171]. The increased expression of miR-221 in psoriasis participates in the regulation of keratinocytes and immune cell proliferation [160]. miR-221 is relevant in controlling the aberrant proliferation of human airway smooth muscle cells in severe asthma [172]. In contrast to the above, miR-125b, responsible for proliferation, is downregulated in psoriatic lesions [21]. In another study, it has been noted that miR-125b in the sputum supernatant was reduced in COPD patients [173].
Another interesting case of post-translational modification concerns sphingosine-1-phosphate receptor-1 (S1PR1), a high-affinity G protein-coupled receptor expressed at the endothelial cell (EC) surface and relevant for the regulation of the immune system [174]. The S1P-S1PR pathway is involved in the pathogenesis of immune-mediated diseases, including psoriasis [175]. The post-translational modification of S1PR1 at serine and tyrosine-143 residues plays a role in disrupting endothelial barrier function, leading to inflammatory conditions of the lungs [176].

4.4.3. Treatment of Psoriasis

Medications used for the treatment of psoriasis can sometimes trigger or exacerbate lung diseases, especially interstitial lung disease. This is an important consideration from the perspective of comorbidity.
Methotrexate, cyclosporine, acitretin, and fumaric acid ester are systemic drugs commonly used in psoriasis. Even though methotrexate [36] and fumaric acid ester [38] have been reported to cause lung fibrosis, this is an extremely rare complication; therefore, all three can be considered generally safe for the respiratory system. Patients treated for psoriatic arthritis with other immunosuppressants, such as leflunomide [39] and sulfasalazine [41], have been reported to develop interstitial lung disease.
Biological medications target specific parts of the immune system. Because they are immunosuppressants, they carry an increased risk of upper respiratory tract infections [177]. According to the latest data, biological drugs used in the treatment of psoriasis probably have a similar risk of serious infections or infections of the respiratory tract [178]. Biologic therapy has been proven to be efficacious for the treatment of psoriasis and psoriatic arthritis. These drugs also affect lung tissue.
Asthma appears to be a definite but rare side effect of the anti-TNF blockade [179]. Moreover, anti-TNF causes exacerbations of asthma, bronchospasm, or wheezing, although most of these have been related to anaphylaxis. This is because it is likely that following the introduction of a TNF-α-blocking drug, the Th1 response was suppressed, allowing the clinical expression of the Th2 pathway as asthma [180]. The appearance of asthma has not been observed with the use of other biological drugs. In the past, anti-TNF-α therapy was viewed as a therapy that caused pulmonary fibrosis. Recent studies have shown that anti-TNF-α therapy does not increase the occurrence of ILD among patients with autoimmune diseases [181].
For the treatment of psoriasis, the following three IL-17-related monoclonal antibodies have been registered: secukinumab, ixekizumab, which directly blocks IL-17A, and brodalumab, which blocks IL-17A receptors [182]. There are no data on the negative effects of anti-IL-17 on lung function. On the contrary, IL-17-neutralizing therapy could be used to treat lung diseases such as asthma, COPD, and possibly fibrosis [183]. Recent studies suggest that asthma may be able to be separated into subtypes: Th2-dominated, Th17-dominated, and Th2/Th17-low, which suggests that αIL-17 therapy may prove effective in the treatment of some types of asthma [184]. Similarly, both COPD and IPF appear to be multidimensional diseases, suggesting that increased levels of IL-17 correlate with more severe disease progression [185,186].
Similar positive conclusions can be drawn from the use of anti-IL-23 in pulmonary disorders in mice models. Treatment with an anti-interleukin-23 antibody attenuated airway inflammation as well as fibrosis and reduced interleukin-17A and -22 levels in a murine model with the exacerbation of pulmonary fibrosis [187]. Treatment with an anti-IL-23p40 monoclonal antibody significantly weakened porcine-pancreatic-elastase-induced emphysematous changes in the lungs of wild-type mice. Targeting IL-23 in emphysema appears to be a therapeutic strategy for delaying disease progression [188].

4.4.4. Underdiagnosis and Treatment of Lung Disease

The underestimated prevalence of lung diseases in psoriasis may result from a mild course of lung disease that remains underdiagnosed. The diagnosis may also be obscured by the presence or current treatment of another concomitant lung disease. An example is pulmonary hypertension, which can develop secondarily into chronic obstructive pulmonary disease, obstructive sleep apnea, sarcoidosis, and pulmonary fibrosis [189]. Whereas all conditions can occur in patients with psoriasis and their prevalence is higher than in the general population (which was discussed above), the presence of COPD, obstructive sleep apnea, sarcoidosis, or pulmonary fibrosis and its current treatment may obscure the clinical features of pulmonary hypertension, thereby leading to its underdiagnosis.

5. Conclusions

Patients with psoriasis have an increased risk of asthma, chronic obstructive pulmonary disease, obstructive sleep apnea, and pulmonary hypertension, which are important, previously unrecognized causes of morbidity and mortality. The development of lung diseases in patients with psoriasis can be explained by several mechanisms, including common risk factors (smoking, obesity, air pollution, infections, exposure to allergens, malnutrition, cardiovascular disease, connective tissue disorder, depression, and use of certain medications), shared immune and molecular characteristics associated with chronic inflammation, and other mechanisms. Understanding the prevalence of lung diseases in psoriasis and their underlying mechanisms can help to develop appropriate preventative and therapeutic strategies to address respiratory diseases in patients with psoriasis.

Author Contributions

Conceptualization, M.M. and D.K.; methodology, M.M.; software, M.M.; validation, M.M., A.G. and D.K.; formal analysis, M.M.; investigation, M.M.; resources, M.M.; data curation, M.M.; writing—original draft preparation, M.M.; writing—review and editing, A.G. and D.K.; visualization, M.M.; supervision, D.K.; project administration, M.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in this article.

Acknowledgments

The authors gratefully acknowledge the assistance of Karolina Kalisz in editing the manuscript.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Boehncke, W.H.; Schön, M.P. Psoriasis. Lancet 2015, 386, 983–994. [Google Scholar] [CrossRef]
  2. Rendon, A.; Schäkel, K. Psoriasis Pathogenesis and Treatment. Int. J. Mol. Sci. 2019, 20, 1475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Liu, J.-T.; Yeh, H.-M.; Liu, S.-Y.; Chen, K.-T. Psoriatic arthritis: Epidemiology, diagnosis, and treatment. World J. Orthop. 2014, 5, 537–543. [Google Scholar] [CrossRef] [PubMed]
  4. Daudén, E.; Castañeda, S.; Suárez, C.; García-Campayo, J.; Blasco, A.J.; Aguilar, M.D.; Ferrándiz, C.; Puig, L.; Sánchez-Carazo, J.L. Clinical practice guideline for an integrated approach to comorbidity in patients with psoriasis. J. Eur. Acad. Dermatol. Venereol. 2013, 27, 1387–1404. [Google Scholar] [CrossRef]
  5. Augustin, M.; Glaeske, G.; Radtke, M.A.; Christophers, E.; Reich, K.; Schäfer, I. Epidemiology and comorbidity of psoriasis in children. Br. J. Dermatol. 2010, 162, 633–636. [Google Scholar] [CrossRef] [PubMed]
  6. Salihbegovic, E.M.; Hadzigrahic, N.; Suljagic, E.; Kurtalic, N.; Hadzic, J.; Zejcirovic, A.; Bijedic, M.; Handanagic, A. Psoriasis and dyslipidemia. Mater. Sociomed. 2015, 27, 15–17. [Google Scholar] [CrossRef] [Green Version]
  7. Armstrong, A.W.; Harskamp, C.T.; Armstrong, E.J. The association between psoriasis and hypertension: A systematic review and meta-analysis of observational studies. J. Hypertens. 2013, 31, 433–443. [Google Scholar] [CrossRef]
  8. Coto-Segura, P.; Eiris-Salvado, N.; González-Lara, L.; Queiro-Silva, R.; Martinez-Camblor, P.; Maldonado-Seral, C.; García-García, B.; Palacios-García, L.; Gomez-Bernal, S.; Santos-Juanes, J.; et al. Psoriasis, psoriatic arthritis and type 2 diabetes mellitus: A systematic review and meta-analysis. Br. J. Dermatol. 2013, 169, 783–793. [Google Scholar] [CrossRef]
  9. Armstrong, A.W.; Harskamp, C.T.; Armstrong, E.J. The association between psoriasis and obesity: A systematic review and meta-analysis of observational studies. Nutr. Diabetes 2012, 2, e54. [Google Scholar] [CrossRef] [Green Version]
  10. Fu, Y.; Lee, C.-H.; Chi, C.-C. Association of Psoriasis with Inflammatory Bowel Disease: A Systematic Review and Meta-analysis. JAMA Dermatol. 2018, 154, 1417–1423. [Google Scholar] [CrossRef] [Green Version]
  11. Ungprasert, P.; Raksasuk, S. Psoriasis and risk of incident chronic kidney disease and end-stage renal disease: A systematic review and meta-analysis. Int. Urol. Nephrol. 2018, 50, 1277–1283. [Google Scholar] [CrossRef] [PubMed]
  12. Wang, J.; Ke, R.; Shi, W.; Yan, X.; Wang, Q.; Zhang, Q.; Chai, L.; Li, M. Association between psoriasis and asthma risk: A meta-analysis. Allergy Asthma Proc. 2018, 39, 103–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Augustin, M.; Radtke, M.A.; Glaeske, G.; Reich, K.; Christophers, E.; Schaefer, I.; Jacobi, A. Epidemiology and Comorbidity in Children with Psoriasis and Atopic Eczema. Dermatology 2015, 231, 35–40. [Google Scholar] [CrossRef] [Green Version]
  14. Fang, H.-Y.; Liao, W.-C.; Lin, C.-L.; Chen, C.-H.; Kao, C.-H. Association between psoriasis and asthma: A population-based retrospective cohort analysis. Br. J. Dermatol. 2015, 172, 1066–1071. [Google Scholar] [CrossRef] [PubMed]
  15. Hajdarbegovic, E.; Nijsten, T.; Westgeest, A.; Habraken, F.; Hollestein, L.; Thio, B. Decreased prevalence of atopic features in patients with psoriatic arthritis, but not in psoriasis vulgaris. J. Am. Acad. Dermatol. 2013, 68, 270–277. [Google Scholar] [CrossRef] [PubMed]
  16. Lønnberg, A.S.; Skov, L.; Skytthe, A.; Kyvik, K.O.; Pedersen, O.B.; Meteran, H.; Backer, V.; Thomsen, S.F. Asthma in patients with psoriasis. Br. J. Dermatol. 2015, 172, 1660–1661. [Google Scholar] [CrossRef] [PubMed]
  17. Galili, E.; Barzilai, A.; Twig, G.; Caspi, T.; Daniely, D.; Shreberk-Hassidim, R.; Astman, N. Allergic Rhinitis and Asthma Among Adolescents with Psoriasis: A Population-based Cross-sectional Study. Acta Derm. Venereol. 2020, 100, adv00133. [Google Scholar] [CrossRef] [PubMed]
  18. Kim, S.Y.; Min, C.; Oh, D.J.; Choi, H.G. Increased risk of psoriasis in children and elderly patients with asthma: A longitudinal follow-up study using a national sample cohort. Int. Forum Allergy Rhinol. 2019, 9, 1304–1310. [Google Scholar] [CrossRef]
  19. Egeberg, A.; Khalid, U.; Gislason, G.H.; Mallbris, L.; Skov, L.; Hansen, P.R. Risk of psoriasis in patients with childhood asthma: A Danish nationwide cohort study. Br. J. Dermatol. 2015, 173, 159–164. [Google Scholar] [CrossRef]
  20. Ungprasert, P.; Srivali, N.; Thongprayoon, C. Association between psoriasis and chronic obstructive pulmonary disease: A systematic review and meta-analysis. J. Dermatolog. Treat. 2016, 27, 316–321. [Google Scholar] [CrossRef]
  21. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Wu, C.-Y.; Hu, H.-Y.; Li, C.-P.; Chou, Y.-J.; Chang, Y.-T. Comorbidity profiles of psoriasis in Taiwan: A latent class analysis. PLoS ONE 2018, 13, e0192537. [Google Scholar] [CrossRef] [Green Version]
  23. Chiang, Y.-Y.; Lin, H.-W. Association between psoriasis and chronic obstructive pulmonary disease: A population-based study in Taiwan. J. Eur. Acad. Dermatol. Venereol. 2012, 26, 59–65. [Google Scholar] [CrossRef]
  24. Al-Mutairi, N.; Al-Farag, S.; Al-Mutairi, A.; Al-Shiltawy, M. Comorbidities associated with psoriasis: An experience from the Middle East. J. Dermatol. 2010, 37, 146–155. [Google Scholar] [CrossRef]
  25. Ger, T.-Y.; Fu, Y.; Chi, C.-C. Bidirectional Association Between Psoriasis and Obstructive Sleep Apnea: A Systematic Review and Meta-Analysis. Sci. Rep. 2020, 10, 5931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Saçmacı, H.; Gürel, G. Sleep disorders in patients with psoriasis: A cross-sectional study using non-polysomnographical methods. Sleep Breath. 2019, 23, 893–898. [Google Scholar] [CrossRef]
  27. Papadavid, E.; Dalamaga, M.; Vlami, K.; Koumaki, D.; Gyftopoulos, S.; Christodoulatos, G.S.; Papiris, S.; Rigopoulos, D. Psoriasis is associated with risk of obstructive sleep apnea independently from metabolic parameters and other comorbidities: A large hospital-based case-control study. Sleep Breath. 2017, 21, 949–958. [Google Scholar] [CrossRef]
  28. Shalom, G.; Dreiher, J.; Cohen, A. Psoriasis and obstructive sleep apnea. Int. J. Dermatol. 2016, 55, e579–e584. [Google Scholar] [CrossRef]
  29. Papadavid, E.; Vlami, K.; Dalamaga, M.; Giatrakou, S.; Theodoropoulos, K.; Gyftopoulos, S.; Stavrianeas, N.; Papiris, S.; Rigopoulos, D. Sleep apnea as a comorbidity in obese psoriasis patients: A cross-sectional study. Do psoriasis characteristics and metabolic parameters play a role? J. Eur. Acad. Dermatol. Venereol. 2013, 27, 820–826. [Google Scholar] [CrossRef]
  30. Yang, Y.-W.; Kang, J.-H.; Lin, H.-C. Increased risk of psoriasis following obstructive sleep apnea: A longitudinal population-based study. Sleep Med. 2012, 13, 285–289. [Google Scholar] [CrossRef]
  31. Choi, Y.M.; Famenini, S.; Wu, J.J. Incidence of Pulmonary Arterial Hypertension in Patients with Psoriasis: A Retrospective Cohort Study. Perm. J. 2017, 21, 16–73. [Google Scholar] [CrossRef] [Green Version]
  32. Ishikawa, G.; Dua, S.; Mathur, A.; Acquah, S.O.; Salvatore, M.; Beasley, M.B.; Padilla, M.L. Concomitant interstitial lung disease with psoriasis. Can. Respir. J. 2019, 2019, 5919304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Gupta, R.; Espiritu, J. Azathioprine for the Rare Case of Nonspecific Interstitial Pneumonitis in a Patient with Psoriasis. Ann. Am. Thorac. Soc. 2015, 12, 1248–1251. [Google Scholar] [CrossRef] [PubMed]
  34. Khalid, U.; Gislason, G.H.; Hansen, P.R. Sarcoidosis in patients with psoriasis: A population-based cohort study. PLoS ONE 2014, 9, e109632. [Google Scholar] [CrossRef] [PubMed]
  35. Egeberg, A.; Khalid, U.; Gislason, G.H.; Mallbris, L.; Skov, L.; Hansen, P.R. Psoriasis and Sleep Apnea: A Danish Nationwide Cohort Study. J. Clin. Sleep Med. 2016, 12, 663–671. [Google Scholar] [CrossRef]
  36. Lateef, O.; Shakoor, N.; Balk, R.A. Methotrexate pulmonary toxicity. Expert Opin. Drug Saf. 2005, 4, 723–730. [Google Scholar] [CrossRef]
  37. Leger, S.; Etienne, M.; Duval-Modeste, A.B.; Roussel, A.; Caron, F.; Thiberville, L. Pneumopathie interstitielle subaiguë aprs traitement d’un psoriasis par infliximab. Ann. Dermatol. Venereol. 2011, 138, 499–503. [Google Scholar] [CrossRef]
  38. Deegan, A.P.; Kirby, B.; Rogers, S.; Crotty, T.B.; McDonnell, T.J. Organising pneumonia associated with fumaric acid ester treatment for psoriasis. Clin. Respir. J. 2010, 4, 248–251. [Google Scholar] [CrossRef]
  39. Lee, M.A.; Hutchinson, D.G. HRCT-proven leflunomide pneumonitis in a patient with psoriatic arthritis and normal lung function tests and chest radiography. Rheumatology 2010, 49, 1206–1207. [Google Scholar] [CrossRef] [Green Version]
  40. Manero Ruiz, F.J.; Larraga Palacio, R.; Herrero Labarga, I.; Ferrer Peralta, M. Pneumonitis caused by gold salts in psoriatic arthritis: Report of 2 cases. An. Med. Interna 2002, 19, 237–240. [Google Scholar]
  41. Woltsche, M.; Woltsche-Kahr, I.; Roeger, G.M.; Aberer, W.; Popper, H. Sulfasalazine-induced extrinsic allergic alveolitis in a patient with psoriatic arthritis. Eur. J. Med. Res. 2001, 6, 495–497. [Google Scholar] [PubMed]
  42. Guzman, L.R.; Gall, E.P.; Pitt, M.; Lull, G. Psoriatic Spondylitis: Association with Advanced Nongranulomatous Upper Lobe Pulmonary Fibrosis. JAMA 1978, 239, 1416–1417. [Google Scholar] [CrossRef] [PubMed]
  43. Osman, S.; Ziegler, C.; Gibson, R.; Mahmood, R.; Moraros, J. The Association between Risk Factors and Chronic Obstructive Pulmonary Disease in Canada: A Cross-sectional Study Using the 2014 Canadian Community Health Survey. Int. J. Prev. Med. 2017, 8, 86. [Google Scholar] [CrossRef] [PubMed]
  44. Cerveri, I.; Cazzoletti, L.; Corsico, A.G.; Marcon, A.; Niniano, R.; Grosso, A.; Ronzoni, V.; Accordini, S.; Janson, C.; Pin, I.; et al. The impact of cigarette smoking on asthma: A population-based international cohort study. Int. Arch. Allergy Immunol. 2012, 158, 175–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Sousa, C.; Rodrigues, M.; Carvalho, A.; Viamonte, B.; Cunha, R.; Guimarães, S.; de Moura, C.S.; Morais, A.; Pereira, J.M. Diffuse smoking-related lung diseases: Insights from a radiologic-pathologic correlation. Insights Imaging 2019, 10, 73. [Google Scholar] [CrossRef] [PubMed]
  46. Fox, B.D.; Azoulay, L.; Dell’Aniello, S.; Langleben, D.; Lapi, F.; Benisty, J.; Suissa, S. The use of antidepressants and the risk of idiopathic pulmonary arterial hypertension. Can. J. Cardiol. 2014, 30, 1633–1639. [Google Scholar] [CrossRef] [PubMed]
  47. Babu, K.S.; Marshall, B.G. Drug-induced airway diseases. Clin. Chest Med. 2004, 25, 113–122. [Google Scholar] [CrossRef]
  48. Ryerson, C.J.; Arean, P.A.; Berkeley, J.; Carrieri-Kohlman, V.L.; Pantilat, S.Z.; Landefeld, C.S.; Collard, H.R. Depression is a common and chronic comorbidity in patients with interstitial lung disease. Respirology 2012, 17, 525–532. [Google Scholar] [CrossRef]
  49. Chaisson, N.F.; Hassoun, P.M. Systemic sclerosis-associated pulmonary arterial hypertension. Chest 2013, 144, 1346–1356. [Google Scholar] [CrossRef] [Green Version]
  50. Perelas, A.; Silver, R.M.; Arrossi, A.V.; Highland, K.B. Systemic sclerosis-associated interstitial lung disease. Lancet Respir. Med. 2020, 8, 304–320. [Google Scholar] [CrossRef]
  51. Fujimura, K.E.; Lynch, S.V. Microbiota in allergy and asthma and the emerging relationship with the gut microbiome. Cell Host Microbe 2015, 17, 592–602. [Google Scholar] [CrossRef] [Green Version]
  52. Kahnert, K.; Lucke, T.; Huber, R.M.; Behr, J.; Biertz, F.; Vogt, A.; Watz, H.; Alter, P.; Fähndrich, S.; Bals, R.; et al. Relationship of hyperlipidemia to comorbidities and lung function in COPD: Results of the COSYCONET cohort. PLoS ONE 2017, 12, e0177501. [Google Scholar] [CrossRef] [PubMed]
  53. Gonçalves, S.C.; Martinez, D.; Gus, M.; de Abreu-Silva, E.O.; Bertoluci, C.; Dutra, I.; Branchi, T.; Moreira, L.B.; Fuchs, S.C.; de Oliveira, A.C.T.; et al. Obstructive sleep apnea and resistant hypertension: A case-control study. Chest 2007, 132, 1858–1862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Ehrlich, S.F.; Quesenberry, C.P.J.; Van Den Eeden, S.K.; Shan, J.; Ferrara, A. Patients diagnosed with diabetes are at increased risk for asthma, chronic obstructive pulmonary disease, pulmonary fibrosis, and pneumonia but not lung cancer. Diabetes Care 2010, 33, 55–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Peters, U.; Dixon, A.E.; Forno, E. Obesity and asthma. J. Allergy Clin. Immunol. 2018, 141, 1169–1179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Badran, M.; Ayas, N.; Laher, I. Insights into obstructive sleep apnea research. Sleep Med. 2014, 15, 485–495. [Google Scholar] [CrossRef] [PubMed]
  57. Peters, U.; Suratt, B.T.; Bates, J.H.T.; Dixon, A.E. Beyond BMI: Obesity and Lung Disease. Chest 2018, 153, 702–709. [Google Scholar] [CrossRef]
  58. Baxi, S.N.; Phipatanakul, W. The role of allergen exposure and avoidance in asthma. Adolesc. Med. State Art Rev. 2010, 21, 57. [Google Scholar]
  59. Crothers, K.; Huang, L.; Goulet, J.L.; Goetz, M.B.; Brown, S.T.; Rodriguez-Barradas, M.C.; Oursler, K.K.; Rimland, D.; Gibert, C.L.; Butt, A.A.; et al. HIV infection and risk for incident pulmonary diseases in the combination antiretroviral therapy era. Am. J. Respir. Crit. Care Med. 2011, 183, 388–395. [Google Scholar] [CrossRef] [Green Version]
  60. Huang, Y.J. The respiratory microbiome and innate immunity in asthma. Curr. Opin. Pulm. Med. 2015, 21, 27–32. [Google Scholar] [CrossRef]
  61. Jarrett, H.; Barnett, C. HIV-associated pulmonary hypertension. Curr. Opin. HIV AIDS 2017, 12, 566–571. [Google Scholar] [CrossRef]
  62. Oh, C.-M.; Oh, I.-H.; Lee, J.-K.; Park, Y.H.; Choe, B.-K.; Yoon, T.-Y.; Choi, J.-M. Blood cadmium levels are associated with a decline in lung function in males. Environ. Res. 2014, 132, 119–125. [Google Scholar] [CrossRef] [PubMed]
  63. Richer, V.; Roubille, C.; Fleming, P.; Starnino, T.; McCourt, C.; McFarlane, A.; Siu, S.; Kraft, J.; Lynde, C.; Pope, J.E.; et al. Psoriasis and Smoking: A Systematic Literature Review and Meta-Analysis with Qualitative Analysis of Effect of Smoking on Psoriasis Severity. J. Cutan. Med. Surg. 2016, 20, 221–227. [Google Scholar] [CrossRef] [PubMed]
  64. Armstrong, A.W.; Armstrong, E.J.; Fuller, E.N.; Sockolov, M.E.; Voyles, S.V. Smoking and pathogenesis of psoriasis: A review of oxidative, inflammatory and genetic mechanisms. Br. J. Dermatol. 2011, 165, 1162–1168. [Google Scholar] [CrossRef] [PubMed]
  65. Fleming, P.; Kraft, J.; Gulliver, W.P.; Lynde, C. The Relationship of Obesity with the Severity of Psoriasis: A Systematic Review. J. Cutan. Med. Surg. 2015, 19, 450–456. [Google Scholar] [CrossRef] [PubMed]
  66. Jacobi, A.; Langenbruch, A.; Purwins, S.; Augustin, M.; Radtke, M.A. Prevalence of Obesity in Patients with Psoriasis: Results of the National Study PsoHealth3. Dermatology 2015, 231, 231–238. [Google Scholar] [CrossRef]
  67. Liaw, F.-Y.; Chen, W.-L.; Kao, T.-W.; Chang, Y.-W.; Huang, C.-F. Exploring the link between cadmium and psoriasis in a nationally representative sample. Sci. Rep. 2017, 7, 1723. [Google Scholar] [CrossRef]
  68. Mallon, E.; Bunker, C.B. HIV-associated psoriasis. AIDS Patient Care STDS 2000, 14, 239–246. [Google Scholar] [CrossRef]
  69. Weryńska-Kalemba, M.; Filipowska-Grońska, A.; Kalemba, M.; Krajewska, A.; Grzanka, A.; Bożek, A.; Jarząb, J. Analysis of selected allergic reactions among psoriatic patients. Postep. Dermatol. Alergol. 2016, 33, 18–22. [Google Scholar] [CrossRef]
  70. Brown, K.; DeCoffe, D.; Molcan, E.; Gibson, D.L. Diet-induced dysbiosis of the intestinal microbiota and the effects on immunity and disease. Nutrients 2012, 4, 1095–1119. [Google Scholar] [CrossRef] [Green Version]
  71. Kuchroo, V.K.; Ohashi, P.S.; Sartor, R.B.; Vinuesa, C.G. Dysregulation of immune homeostasis in autoimmune diseases. Nat. Med. 2012, 18, 42–47. [Google Scholar] [CrossRef] [PubMed]
  72. Gunawan, H.; Awalia, A.; Soeroso, J. The Coexistence of Systemic Lupus Erythematosus and Psoriasis: Is It Possible? Acta Med. Indones. 2018, 50, 144–150. [Google Scholar] [PubMed]
  73. Watad, A.; Bragazzi, N.L.; McGonagle, D.; Damiani, G.; Comaneshter, D.; Cohen, A.; Amital, H. Systemic Sclerosis is Linked to Psoriasis and May Impact on Patients’ Survival: A Large Cohort Study. J. Clin. Med. 2019, 8, 521. [Google Scholar] [CrossRef] [Green Version]
  74. García-Sánchez, L.; Montiel-Jarquín, Á.J.; Vázquez-Cruz, E.; May-Salazar, A.; Gutiérrez-Gabriel, I.; Loría-Castellanoso, J. Quality of life in patients with psoriasis. Gac. Med. Mex. 2017, 153, 185–189. [Google Scholar]
  75. Snast, I.; Reiter, O.; Atzmony, L.; Leshem, Y.A.; Hodak, E.; Mimouni, D.; Pavlovsky, L. Psychological stress and psoriasis: A systematic review and meta-analysis. Br. J. Dermatol. 2018, 178, 1044–1055. [Google Scholar] [CrossRef]
  76. Dowlati, Y.; Herrmann, N.; Swardfager, W.; Liu, H.; Sham, L.; Reim, E.K.; Lanctôt, K.L. A meta-analysis of cytokines in major depression. Biol. Psychiatry 2010, 67, 446–457. [Google Scholar] [CrossRef] [PubMed]
  77. Balak, D.M.; Hajdarbegovic, E. Drug-induced psoriasis: Clinical perspectives. Psoriasis 2017, 7, 87–94. [Google Scholar] [CrossRef] [Green Version]
  78. Wee, J.S.; Natkunarajah, J.; Moosa, Y.; Marsden, R.A. Erythrodermic pustular psoriasis triggered by intravesical bacillus Calmette-Guérin immunotherapy. Clin. Exp. Dermatol. 2012, 37, 455–457. [Google Scholar] [CrossRef]
  79. Rogoziński, P.; Taracha-Guz, D.; Pęcikiewicz, P.; Kachel, T.; Dubiel, G.; Wandzel, P.; Bruliński, K. Granulomatous pneumonia as a complication of intravesical BCG immunotherapy—A case report. Pneumonol. Alergol. Pol. 2014, 82, 163–169. [Google Scholar] [CrossRef] [Green Version]
  80. Reich, K. The concept of psoriasis as a systemic inflammation: Implications for disease management. J. Eur. Acad. Dermatol. Venereol. 2012, 26 (Suppl. S2), 3–11. [Google Scholar] [CrossRef]
  81. Robinson, D.J.; Hackett, M.; Wong, J.; Kimball, A.B.; Cohen, R.; Bala, M. Co-occurrence and comorbidities in patients with immune-mediated inflammatory disorders: An exploration using US healthcare claims data, 2001–2002. Curr. Med. Res. Opin. 2006, 22, 989–1000. [Google Scholar] [CrossRef] [PubMed]
  82. Suárez-Fariñas, M.; Li, K.; Fuentes-Duculan, J.; Hayden, K.; Brodmerkel, C.; Krueger, J.G. Expanding the psoriasis disease profile: Interrogation of the skin and serum of patients with moderate-to-severe psoriasis. J. Investig. Dermatol. 2012, 132, 2552–2564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Jin, Y.; Wan, Y.; Chen, G.; Chen, L.; Zhang, M.-Q.; Deng, L.; Zhang, J.-C.; Xiong, X.-Z.; Xin, J.-B. Treg/IL-17 ratio and Treg differentiation in patients with COPD. PLoS ONE 2014, 9, e111044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Liu, Y.; Krueger, J.G.; Bowcock, A.M. Psoriasis: Genetic associations and immune system changes. Genes Immun. 2007, 8, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Lowes, M.A.; Bowcock, A.M.; Krueger, J.G. Pathogenesis and therapy of psoriasis. Nature 2007, 445, 866–873. [Google Scholar] [CrossRef]
  86. Griffiths, C.E.M.; Iaccarino, L.; Naldi, L.; Olivieri, I.; Pipitone, N.; Salvarani, C.; Doria, A. Psoriasis and psoriatic arthritis: Immunological aspects and therapeutic guidelines. Clin. Exp. Rheumatol. 2006, 24, S72–S78. [Google Scholar]
  87. Griffiths, C.E.; Barker, J.N. Pathogenesis and clinical features of psoriasis. Lancet 2007, 370, 263–271. [Google Scholar] [CrossRef]
  88. Maari, C.; Bolduc, C.; Nigen, S.; Marchessault, P.; Bissonnette, R. Effect of adalimumab on sleep parameters in patients with psoriasis and obstructive sleep apnea: A randomized controlled trial. J. Dermatolog. Treat. 2014, 25, 57–60. [Google Scholar] [CrossRef]
  89. Diani, M.; Altomare, G.; Reali, E. T Helper Cell Subsets in Clinical Manifestations of Psoriasis. J. Immunol. Res. 2016, 2016, 7692024. [Google Scholar] [CrossRef] [Green Version]
  90. Chang, H.S.; Lee, T.-H.; Jun, J.A.; Baek, A.R.; Park, J.-S.; Koo, S.-M.; Kim, Y.-K.; Lee, H.S.; Park, C.-S. Neutrophilic inflammation in asthma: Mechanisms and therapeutic considerations. Expert Rev. Respir. Med. 2017, 11, 29–40. [Google Scholar] [CrossRef]
  91. Urbanowicz, R.A.; Lamb, J.R.; Todd, I.; Corne, J.M.; Fairclough, L.C. Enhanced effector function of cytotoxic cells in the induced sputum of COPD patients. Respir. Res. 2010, 11, 76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Di Stefano, A.; Caramori, G.; Gnemmi, I.; Contoli, M.; Vicari, C.; Capelli, A.; Magno, F.; D’Anna, S.E.; Zanini, A.; Brun, P.; et al. T helper type 17-related cytokine expression is increased in the bronchial mucosa of stable chronic obstructive pulmonary disease patients. Clin. Exp. Immunol. 2009, 157, 316–324. [Google Scholar] [CrossRef] [PubMed]
  93. Facco, M.; Cabrelle, A.; Teramo, A.; Olivieri, V.; Gnoato, M.; Teolato, S.; Ave, E.; Gattazzo, C.; Fadini, G.P.; Calabrese, F.; et al. Sarcoidosis is a Th1/Th17 multisystem disorder. Thorax 2011, 66, 144–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Molet, S.; Hamid, Q.; Davoine, F.; Nutku, E.; Taha, R.; Pagé, N.; Olivenstein, R.; Elias, J.; Chakir, J. IL-17 is increased in asthmatic airways and induces human bronchial fibroblasts to produce cytokines. J. Allergy Clin. Immunol. 2001, 108, 430–438. [Google Scholar] [CrossRef] [PubMed]
  95. Raphael, I.; Nalawade, S.; Eagar, T.N.; Forsthuber, T.G. T cell subsets and their signature cytokines in autoimmune and inflammatory diseases. Cytokine 2015, 74, 5–17. [Google Scholar] [CrossRef] [Green Version]
  96. Nickoloff, B.J.; Nestle, F.O. Recent insights into the immunopathogenesis of psoriasis provide new therapeutic opportunities. J. Clin. Investig. 2004, 113, 1664–1675. [Google Scholar] [CrossRef]
  97. Eurlings, I.M.J.; Dentener, M.A.; Mercken, E.M.; de Cabo, R.; Bracke, K.R.; Vernooy, J.H.J.; Wouters, E.F.M.; Reynaert, N.L. A comparative study of matrix remodeling in chronic models for COPD; mechanistic insights into the role of TNF-α. Am. J. Physiol. Lung Cell. Mol. Physiol. 2014, 307, L557–L565. [Google Scholar] [CrossRef] [Green Version]
  98. Chung, K.F.; Adcock, I.M. Multifaceted mechanisms in COPD: Inflammation, immunity, and tissue repair and destruction. Eur. Respir. J. 2008, 31, 1334–1356. [Google Scholar] [CrossRef]
  99. Entzian, P.; Linnemann, K.; Schlaak, M.; Zabel, P. Obstructive sleep apnea syndrome and circadian rhythms of hormones and cytokines. Am. J. Respir. Crit. Care Med. 1996, 153, 1080–1086. [Google Scholar] [CrossRef]
  100. Carpagnano, G.E.; Kharitonov, S.A.; Resta, O.; Foschino-Barbaro, M.P.; Gramiccioni, E.; Barnes, P.J. Increased 8-isoprostane and interleukin-6 in breath condensate of obstructive sleep apnea patients. Chest 2002, 122, 1162–1167. [Google Scholar] [CrossRef]
  101. Wang, Z.; Zheng, T.; Zhu, Z.; Homer, R.J.; Riese, R.J.; Chapman, H.A.J.; Shapiro, S.D.; Elias, J.A. Interferon gamma induction of pulmonary emphysema in the adult murine lung. J. Exp. Med. 2000, 192, 1587–1600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Churg, A.; Wang, R.D.; Tai, H.; Wang, X.; Xie, C.; Wright, J.L. Tumor necrosis factor-alpha drives 70% of cigarette smoke-induced emphysema in the mouse. Am. J. Respir. Crit. Care Med. 2004, 170, 492–498. [Google Scholar] [CrossRef] [PubMed]
  103. Shim, J.J.; Dabbagh, K.; Ueki, I.F.; Dao-Pick, T.; Burgel, P.R.; Takeyama, K.; Tam, D.C.; Nadel, J.A. IL-13 induces mucin production by stimulating epidermal growth factor receptors and by activating neutrophils. Am. J. Physiol. Lung Cell. Mol. Physiol. 2001, 280, L134–L140. [Google Scholar] [CrossRef] [PubMed]
  104. Singh, T.P.; Schön, M.P.; Wallbrecht, K.; Gruber-Wackernagel, A.; Wang, X.J.; Wolf, P. Involvement of IL-9 in Th17-Associated Inflammation and Angiogenesis of Psoriasis. PLoS ONE 2013, 8, e51752. [Google Scholar] [CrossRef]
  105. Bullens, D.M.A.; Truyen, E.; Coteur, L.; Dilissen, E.; Hellings, P.W.; Dupont, L.J.; Ceuppens, J.L. IL-17 mRNA in sputum of asthmatic patients: Linking T cell driven inflammation and granulocytic influx? Respir. Res. 2006, 7, 135. [Google Scholar] [CrossRef] [Green Version]
  106. Ten Berge, B.; Paats, M.S.; Bergen, I.M.; van den Blink, B.; Hoogsteden, H.C.; Lambrecht, B.N.; Hendriks, R.W.; Kleinjan, A. Increased IL-17A expression in granulomas and in circulating memory T cells in sarcoidosis. Rheumatology 2012, 51, 37–46. [Google Scholar] [CrossRef] [Green Version]
  107. Ryan, S.; Taylor, C.T.; McNicholas, W.T. Selective activation of inflammatory pathways by intermittent hypoxia in obstructive sleep apnea syndrome. Circulation 2005, 112, 2660–2667. [Google Scholar] [CrossRef] [Green Version]
  108. Minoguchi, K.; Tazaki, T.; Yokoe, T.; Minoguchi, H.; Watanabe, Y.; Yamamoto, M.; Adachi, M. Elevated production of tumor necrosis factor-alpha by monocytes in patients with obstructive sleep apnea syndrome. Chest 2004, 126, 1473–1479. [Google Scholar] [CrossRef]
  109. Yue, H.J.; Mills, P.J.; Ancoli-Israel, S.; Loredo, J.S.; Ziegler, M.G.; Dimsdale, J.E. The roles of TNF-alpha and the soluble TNF receptor I on sleep architecture in OSA. Sleep Breath 2009, 13, 263–269. [Google Scholar] [CrossRef] [Green Version]
  110. Östling, J.; van Geest, M.; Schofield, J.P.R.; Jevnikar, Z.; Wilson, S.; Ward, J.; Lutter, R.; Shaw, D.E.; Bakke, P.S.; Caruso, M.; et al. IL-17-high asthma with features of a psoriasis immunophenotype. J. Allergy Clin. Immunol. 2019, 144, 1198–1213. [Google Scholar] [CrossRef] [Green Version]
  111. Park, H.; Li, Z.; Yang, X.O.; Chang, S.H.; Nurieva, R.; Wang, Y.-H.; Wang, Y.; Hood, L.; Zhu, Z.; Tian, Q.; et al. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat. Immunol. 2005, 6, 1133–1141. [Google Scholar] [CrossRef] [PubMed]
  112. Yang, X.O.; Chang, S.H.; Park, H.; Nurieva, R.; Shah, B.; Acero, L.; Wang, Y.-H.; Schluns, K.S.; Broaddus, R.R.; Zhu, Z.; et al. Regulation of inflammatory responses by IL-17F. J. Exp. Med. 2008, 205, 1063–1075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Jia, L.; Wu, C. The biology and functions of Th22 cells. Adv. Exp. Med. Biol. 2014, 841, 209–230. [Google Scholar] [CrossRef] [PubMed]
  114. Nograles, K.E.; Zaba, L.C.; Guttman-Yassky, E.; Fuentes-Duculan, J.; Suárez-Fariñas, M.; Cardinale, I.; Khatcherian, A.; Gonzalez, J.; Pierson, K.C.; White, T.R.; et al. Th17 cytokines interleukin (IL)-17 and IL-22 modulate distinct inflammatory and keratinocyte-response pathways. Br. J. Dermatol. 2008, 159, 1092–1102. [Google Scholar] [CrossRef] [Green Version]
  115. Wolk, K.; Witte, E.; Wallace, E.; Döcke, W.-D.; Kunz, S.; Asadullah, K.; Volk, H.-D.; Sterry, W.; Sabat, R. IL-22 regulates the expression of genes responsible for antimicrobial defense, cellular differentiation, and mobility in keratinocytes: A potential role in psoriasis. Eur. J. Immunol. 2006, 36, 1309–1323. [Google Scholar] [CrossRef]
  116. Whittington, H.A.; Armstrong, L.; Uppington, K.M.; Millar, A.B. Interleukin-22: A potential immunomodulatory molecule in the lung. Am. J. Respir. Cell Mol. Biol. 2004, 31, 220–226. [Google Scholar] [CrossRef]
  117. Starkey, M.R.; Plank, M.W.; Casolari, P.; Papi, A.; Pavlidis, S.; Guo, Y.; Cameron, G.J.M.; Haw, T.J.; Tam, A.; Obiedat, M.; et al. IL-22 and its receptors are increased in human and experimental COPD and contribute to pathogenesis. Eur. Respir. J. 2019, 54, 1800174. [Google Scholar] [CrossRef]
  118. Ziegenhagen, M.W.; Müller-Quernheim, J. The cytokine network in sarcoidosis and its clinical relevance. J. Intern. Med. 2003, 253, 18–30. [Google Scholar] [CrossRef] [Green Version]
  119. Kaplan, M.H. Th9 cells: Differentiation and disease. Immunol. Rev. 2013, 252, 104–115. [Google Scholar] [CrossRef]
  120. Schlapbach, C.; Gehad, A.; Yang, C.; Watanabe, R.; Guenova, E.; Teague, J.E.; Campbell, L.; Yawalkar, N.; Kupper, T.S.; Clark, R.A. Human TH9 cells are skin-tropic and have autocrine and paracrine proinflammatory capacity. Sci. Transl. Med. 2014, 6, 219ra8. [Google Scholar] [CrossRef] [Green Version]
  121. Soroosh, P.; Doherty, T.A. Th9 and allergic disease. Immunology 2009, 127, 450–458. [Google Scholar] [CrossRef] [PubMed]
  122. Lapperre, T.S.; Postma, D.S.; Gosman, M.M.E.; Snoeck-Stroband, J.B.; ten Hacken, N.H.T.; Hiemstra, P.S.; Timens, W.; Sterk, P.J.; Mauad, T. Relation between duration of smoking cessation and bronchial inflammation in COPD. Thorax 2006, 61, 115–121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Huang, N.; Liu, L.; Wang, X.-Z.; Liu, D.; Yin, S.-Y.; Yang, X.-D. Association of interleukin (IL)-12 and IL-27 gene polymorphisms with chronic obstructive pulmonary disease in a Chinese population. DNA Cell Biol. 2008, 27, 527–531. [Google Scholar] [CrossRef] [PubMed]
  124. Zhang, L.; Cheng, Z.; Liu, W.; Wu, K. Expression of interleukin (IL)-10, IL-17A and IL-22 in serum and sputum of stable chronic obstructive pulmonary disease patients. COPD 2013, 10, 459–465. [Google Scholar] [CrossRef] [PubMed]
  125. Flisiak, I.; Zaniewski, P.; Chodynicka, B. Plasma TGF-beta1, TIMP-1, MMP-1 and IL-18 as a combined biomarker of psoriasis activity. Biomarkers 2008, 13, 549–556. [Google Scholar] [CrossRef] [PubMed]
  126. Zaba, L.C.; Fuentes-Duculan, J.; Eungdamrong, N.J.; Johnson-Huang, L.M.; Nograles, K.E.; White, T.R.; Pierson, K.C.; Lentini, T.; Suárez-Fariñas, M.; Lowes, M.A.; et al. Identification of TNF-related apoptosis-inducing ligand and other molecules that distinguish inflammatory from resident dendritic cells in patients with psoriasis. J. Allergy Clin. Immunol. 2010, 125, 1261–1268.e9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Gabryelska, A.; Szmyd, B.; Panek, M.; Szemraj, J.; Kuna, P.; Białasiewicz, P. Serum hypoxia-inducible factor-1α protein level as a diagnostic marker of obstructive sleep apnea. Pol. Arch. Intern. Med. 2020, 130, 158–160. [Google Scholar] [CrossRef] [Green Version]
  128. Kaczmarek, E.; Bakker, J.P.; Clarke, D.N.; Csizmadia, E.; Kocher, O.; Veves, A.; Tecilazich, F.; O’Donnell, C.P.; Ferran, C.; Malhotra, A. Molecular biomarkers of vascular dysfunction in obstructive sleep apnea. PLoS ONE 2013, 8, e70559. [Google Scholar] [CrossRef] [Green Version]
  129. Kondo, S.; Yoneta, A.; Yazawa, H.; Kamada, A.; Jimbow, K. Downregulation of CXCR-2 but not CXCR-1 expression by human keratinocytes by UVB. J. Cell. Physiol. 2000, 182, 366–370. [Google Scholar] [CrossRef]
  130. Singh, K.; Gatzka, M.; Peters, T.; Borkner, L.; Hainzl, A.; Wang, H.; Sindrilaru, A.; Scharffetter-Kochanek, K. Reduced CD18 levels drive regulatory T cell conversion into Th17 cells in the CD18hypo PL/J mouse model of psoriasis. J. Immunol. 2013, 190, 2544–2553. [Google Scholar] [CrossRef] [Green Version]
  131. Starodubtseva, N.L.; Sobolev, V.V.; Soboleva, A.G.; Nikolaev, A.A.; Bruskin, S.A. Expression of genes for metalloproteinases (MMP-1, MMP-2, MMP-9, and MMP-12) associated with psoriasis. Genetika 2011, 47, 1254–1261. [Google Scholar] [CrossRef] [PubMed]
  132. Elder, J.T. Genome-wide association scan yields new insights into the immunopathogenesis of psoriasis. Genes Immun. 2009, 10, 201–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Weidinger, S.; Willis-Owen, S.A.G.; Kamatani, Y.; Baurecht, H.; Morar, N.; Liang, L.; Edser, P.; Street, T.; Rodriguez, E.; O’Regan, G.M.; et al. A genome-wide association study of atopic dermatitis identifies loci with overlapping effects on asthma and psoriasis. Hum. Mol. Genet. 2013, 22, 4841–4856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Kere, J. Mapping and identifying genes for asthma and psoriasis. Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci. 2005, 360, 1551–1561. [Google Scholar] [CrossRef] [Green Version]
  135. Hollox, E.J.; Huffmeier, U.; Zeeuwen, P.L.J.M.; Palla, R.; Lascorz, J.; Rodijk-Olthuis, D.; van de Kerkhof, P.C.M.; Traupe, H.; de Jongh, G.; den Heijer, M.; et al. Psoriasis is associated with increased beta-defensin genomic copy number. Nat. Genet. 2008, 40, 23–25. [Google Scholar] [CrossRef] [Green Version]
  136. Janssens, W.; Nuytten, H.; Dupont, L.J.; Van Eldere, J.; Vermeire, S.; Lambrechts, D.; Nackaerts, K.; Decramer, M.; Cassiman, J.-J.; Cuppens, H. Genomic copy number determines functional expression of {beta}-defensin 2 in airway epithelial cells and associates with chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2010, 182, 163–169. [Google Scholar] [CrossRef]
  137. Huang, S.L.; Su, C.H.; Chang, S.C. Tumor necrosis factor-alpha gene polymorphism in chronic bronchitis. Am. J. Respir. Crit. Care Med. 1997, 156, 1436–1439. [Google Scholar] [CrossRef]
  138. Balding, J.; Kane, D.; Livingstone, W.; Mynett-Johnson, L.; Bresnihan, B.; Smith, O.; FitzGerald, O. Cytokine gene polymorphisms: Association with psoriatic arthritis susceptibility and severity. Arthritis Rheum. 2003, 48, 1408–1413. [Google Scholar] [CrossRef]
  139. Nair, R.P.; Duffin, K.C.; Helms, C.; Ding, J.; Stuart, P.E.; Goldgar, D.; Gudjonsson, J.E.; Li, Y.; Tejasvi, T.; Feng, B.-J.; et al. Genome-wide scan reveals association of psoriasis with IL-23 and NF-kappaB pathways. Nat. Genet. 2009, 41, 199–204. [Google Scholar] [CrossRef] [Green Version]
  140. Kim, H.S.; Choi, D.; Lim, L.L.; Allada, G.; Smith, J.R.; Austin, C.R.; Doyle, T.M.; Goodwin, K.A.; Rosenbaum, J.T.; Martin, T.M. Association of interleukin 23 receptor gene with sarcoidosis. Dis. Markers 2011, 31, 17–24. [Google Scholar] [CrossRef]
  141. Shen, Z. Genomic instability and cancer: An introduction. J. Mol. Cell Biol. 2011, 3, 1–13. [Google Scholar] [CrossRef] [PubMed]
  142. Karaman, A.; Aliağaoğlu, C.; Pirim, I. Sister chromatid exchange analysis in patients with psoriasis. Exp. Dermatol. 2008, 17, 524–529. [Google Scholar] [CrossRef] [PubMed]
  143. Casella, M.; Miniati, M.; Monti, S.; Minichilli, F.; Bianchi, F.; Simi, S. No evidence of chromosome damage in chronic obstructive pulmonary disease (COPD). Mutagenesis 2006, 21, 167–171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Paraskakis, E.; Sourvinos, G.; Passam, F.; Tzanakis, N.; Tzortzaki, E.G.; Zervou, M.; Spandidos, D.; Siafakas, N.M. Microsatellite DNA instability and loss of heterozygosity in bronchial asthma. Eur. Respir. J. 2003, 22, 951–955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Spandidos, D.; Ergazaki, M.; Hatzistamou, J.; Kiaris, H.; Bouros, D.; Tzortzaki, E.; Siafakas, N. Microsatellite instability in patients with chronic obstructive pulmonary disease. Oncol. Rep. 1996, 3, 489–491. [Google Scholar] [CrossRef] [Green Version]
  146. Vassilakis, D.A.; Sourvinos, G.; Markatos, M.; Psathakis, K.; Spandidos, D.A.; Siafakas, N.M.; Bouros, D. Microsatellite DNA instability and loss of heterozygosity in pulmonary sarcoidosis. Am. J. Respir. Crit. Care Med. 1999, 160, 1729–1733. [Google Scholar] [CrossRef]
  147. Demopoulos, K.; Arvanitis, D.A.; Vassilakis, D.A.; Siafakas, N.M.; Spandidos, D.A. MYCL1, FHIT, SPARC, p16(INK4) and TP53 genes associated to lung cancer in idiopathic pulmonary fibrosis. J. Cell. Mol. Med. 2002, 6, 215–222. [Google Scholar] [CrossRef]
  148. Zachos, G.; Koumantaki, E.; Vareltzidis, A.; Spandidos, D.A. Evidence for loss of heterozygosity in human psoriatic lesions. Br. J. Dermatol. 1998, 139, 974–977. [Google Scholar] [CrossRef]
  149. Hyun, J.S.; Jo, B.-K.; Park, C.J.; Yi, J.Y.; Lee, J.Y.; Rhyu, M.-G. Loss of heterozygosity and PCR artifacts in a microsatellite analysis of psoriasis and colorectal cancer. J. Korean Med. Sci. 2002, 17, 641–647. [Google Scholar] [CrossRef] [Green Version]
  150. Anderson, G.P.; Bozinovski, S. Acquired somatic mutations in the molecular pathogenesis of COPD. Trends Pharmacol. Sci. 2003, 24, 71–76. [Google Scholar] [CrossRef]
  151. Anthonisen, N.R.; Skeans, M.A.; Wise, R.A.; Manfreda, J.; Kanner, R.E.; Connett, J.E. The effects of a smoking cessation intervention on 14.5-year mortality: A randomized clinical trial. Ann. Intern. Med. 2005, 142, 233–239. [Google Scholar] [CrossRef] [PubMed]
  152. Shao, S.; Gudjonsson, J.E. Epigenetics of Psoriasis. Adv. Exp. Med. Biol. 2020, 1253, 209–221. [Google Scholar] [CrossRef] [PubMed]
  153. Dopytalska, K.; Ciechanowicz, P.; Wiszniewski, K.; Szymańska, E.; Walecka, I. The Role of Epigenetic Factors in Psoriasis. Int. J. Mol. Sci. 2021, 22, 9294. [Google Scholar] [CrossRef] [PubMed]
  154. Zhang, P.; Su, Y.; Zhao, M.; Huang, W.; Lu, Q. Abnormal histone modifications in PBMCs from patients with psoriasis vulgaris. Eur. J. Dermatol. 2011, 21, 552–557. [Google Scholar] [CrossRef] [PubMed]
  155. Sanders, Y.Y.; Tollefsbol, T.O.; Varisco, B.M.; Hagood, J.S. Epigenetic regulation of thy-1 by histone deacetylase inhibitor in rat lung fibroblasts. Am. J. Respir. Cell Mol. Biol. 2011, 45, 16–23. [Google Scholar] [CrossRef] [Green Version]
  156. Ding, J.; Li, F.; Cong, Y.; Miao, J.; Wu, D.; Liu, B.; Wang, L. Trichostatin A inhibits skeletal muscle atrophy induced by cigarette smoke exposure in mice. Life Sci. 2019, 235, 116800. [Google Scholar] [CrossRef]
  157. Ovejero-Benito, M.C.; Reolid, A.; Sánchez-Jiménez, P.; Saiz-Rodríguez, M.; Muñoz-Aceituno, E.; Llamas-Velasco, M.; Martín-Vilchez, S.; Cabaleiro, T.; Román, M.; Ochoa, D.; et al. Histone modifications associated with biological drug response in moderate-to-severe psoriasis. Exp. Dermatol. 2018, 27, 1361–1371. [Google Scholar] [CrossRef]
  158. Seumois, G.; Chavez, L.; Gerasimova, A.; Lienhard, M.; Omran, N.; Kalinke, L.; Vedanayagam, M.; Ganesan, A.P.V.; Chawla, A.; Djukanović, R.; et al. Epigenomic analysis of primary human T cells reveals enhancers associated with TH2 memory cell differentiation and asthma susceptibility. Nat. Immunol. 2014, 15, 777–788. [Google Scholar] [CrossRef]
  159. Andresen, E.; Günther, G.; Bullwinkel, J.; Lange, C.; Heine, H. Increased expression of beta-defensin 1 (DEFB1) in chronic obstructive pulmonary disease. PLoS ONE 2011, 6, e21898. [Google Scholar] [CrossRef]
  160. Tsoi, L.C.; Iyer, M.K.; Stuart, P.E.; Swindell, W.R.; Gudjonsson, J.E.; Tejasvi, T.; Sarkar, M.K.; Li, B.; Ding, J.; Voorhees, J.J.; et al. Analysis of long non-coding RNAs highlights tissue-specific expression patterns and epigenetic profiles in normal and psoriatic skin. Genome Biol. 2015, 16, 24. [Google Scholar] [CrossRef] [Green Version]
  161. Hermann, H.; Runnel, T.; Aab, A.; Baurecht, H.; Rodriguez, E.; Magilnick, N.; Urgard, E.; Šahmatova, L.; Prans, E.; Maslovskaja, J.; et al. miR-146b Probably Assists miRNA-146a in the Suppression of Keratinocyte Proliferation and Inflammatory Responses in Psoriasis. J. Investig. Dermatol. 2017, 137, 1945–1954. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Comer, B.S.; Camoretti-Mercado, B.; Kogut, P.C.; Halayko, A.J.; Solway, J.; Gerthoffer, W.T. MicroRNA-146a and microRNA-146b expression and anti-inflammatory function in human airway smooth muscle. Am. J. Physiol. Lung Cell. Mol. Physiol. 2014, 307, L727–L734. [Google Scholar] [CrossRef] [PubMed]
  163. Garbacki, N.; Di Valentin, E.; Huynh-Thu, V.A.; Geurts, P.; Irrthum, A.; Crahay, C.; Arnould, T.; Deroanne, C.; Piette, J.; Cataldo, D.; et al. MicroRNAs profiling in murine models of acute and chronic asthma: A relationship with mRNAs targets. PLoS ONE 2011, 6, e16509. [Google Scholar] [CrossRef] [Green Version]
  164. Huang, R.-Y.; Li, L.; Wang, M.-J.; Chen, X.-M.; Huang, Q.-C.; Lu, C.-J. An Exploration of the Role of MicroRNAs in Psoriasis: A Systematic Review of the Literature. Medicine 2015, 94, e2030. [Google Scholar] [CrossRef] [PubMed]
  165. Singhvi, G.; Manchanda, P.; Krishna Rapalli, V.; Kumar Dubey, S.; Gupta, G.; Dua, K. MicroRNAs as biological regulators in skin disorders. Biomed. Pharmacother. 2018, 108, 996–1004. [Google Scholar] [CrossRef]
  166. Sun, W.; Xiao, B.; Jia, A.; Qiu, L.; Zeng, Q.; Liu, D.; Yuan, Y.; Jia, J.; Zhang, X.; Xiang, X. MBD2-mediated Th17 differentiation in severe asthma is associated with impaired SOCS3 expression. Exp. Cell Res. 2018, 371, 196–204. [Google Scholar] [CrossRef] [PubMed]
  167. Dong, R.; Xie, L.; Zhao, K.; Zhang, Q.; Zhou, M.; He, P. Cigarette smoke-induced lung inflammation in COPD mediated via LTB4/BLT1/SOCS1 pathway. Int. J. Chron. Obstruct. Pulmon. Dis. 2016, 11, 31–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Meng, Z.; Zhou, D.; Gao, Y.; Zeng, M.; Wang, W. miRNA delivery for skin wound healing. Adv. Drug Deliv. Rev. 2018, 129, 308–318. [Google Scholar] [CrossRef]
  169. Lu, T.X.; Munitz, A.; Rothenberg, M.E. MicroRNA-21 is up-regulated in allergic airway inflammation and regulates IL-12p35 expression. J. Immunol. 2009, 182, 4994–5002. [Google Scholar] [CrossRef] [Green Version]
  170. Yang, S.; Banerjee, S.; de Freitas, A.; Sanders, Y.Y.; Ding, Q.; Matalon, S.; Thannickal, V.J.; Abraham, E.; Liu, G. Participation of miR-200 in pulmonary fibrosis. Am. J. Pathol. 2012, 180, 484–493. [Google Scholar] [CrossRef] [Green Version]
  171. Li, Y.; Huang, J.; Hu, C.; Zhou, J.; Xu, D.; Hou, Y.; Wu, C.; Zhao, J.; Li, M.; Zeng, X.; et al. MicroRNA-320a: An important regulator in the fibrotic process in interstitial lung disease of systemic sclerosis. Arthritis Res. Ther. 2021, 23, 21. [Google Scholar] [CrossRef] [PubMed]
  172. Perry, M.M.; Baker, J.E.; Gibeon, D.S.; Adcock, I.M.; Chung, K.F. Airway smooth muscle hyperproliferation is regulated by microRNA-221 in severe asthma. Am. J. Respir. Cell Mol. Biol. 2014, 50, 7–17. [Google Scholar] [CrossRef] [Green Version]
  173. Van Pottelberge, G.R.; Mestdagh, P.; Bracke, K.R.; Thas, O.; van Durme, Y.M.T.A.; Joos, G.F.; Vandesompele, J.; Brusselle, G.G. MicroRNA expression in induced sputum of smokers and patients with chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2011, 183, 898–906. [Google Scholar] [CrossRef]
  174. Peyrin-Biroulet, L.; Christopher, R.; Behan, D.; Lassen, C. Modulation of sphingosine-1-phosphate in inflammatory bowel disease. Autoimmun. Rev. 2017, 16, 495–503. [Google Scholar] [CrossRef] [PubMed]
  175. Pérez-Jeldres, T.; Alvarez-Lobos, M.; Rivera-Nieves, J. Targeting Sphingosine-1-Phosphate Signaling in Immune-Mediated Diseases: Beyond Multiple Sclerosis. Drugs 2021, 81, 985–1002. [Google Scholar] [CrossRef] [PubMed]
  176. Anwar, M.; Mehta, D. Post-translational modifications of S1PR1 and endothelial barrier regulation. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2020, 1865, 158760. [Google Scholar] [CrossRef] [PubMed]
  177. Kamata, M.; Tada, Y. Efficacy and Safety of Biologics for Psoriasis and Psoriatic Arthritis and Their Impact on Comorbidities: A Literature Review. Int. J. Mol. Sci. 2020, 21, 1690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Yiu, Z.Z.N.; Smith, C.H.; Ashcroft, D.M.; Lunt, M.; Walton, S.; Murphy, R.; Reynolds, N.J.; Ormerod, A.D.; Griffiths, C.E.M.; Warren, R.B. Risk of Serious Infection in Patients with Psoriasis Receiving Biologic Therapies: A Prospective Cohort Study from the British Association of Dermatologists Biologic Interventions Register (BADBIR). J. Investig. Dermatol. 2018, 138, 534–541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Eren Dagli, C.; Akgedik, R.; Yildirim, B.B.; Gunaydin, R. Adalimumab and etanercept induced asthma. Eur. Respir. J. 2015, 46, PA650. [Google Scholar] [CrossRef]
  180. Bennett, A.N.; Wong, M.; Zain, A.; Panayi, G.; Kirkham, B. Adalimumab-induced asthma. Rheumatology 2005, 44, 1199–1200. [Google Scholar] [CrossRef] [Green Version]
  181. Herrinton, L.J.; Harrold, L.R.; Liu, L.; Raebel, M.A.; Taharka, A.; Winthrop, K.L.; Solomon, D.H.; Curtis, J.R.; Lewis, J.D.; Saag, K.G. Association between anti-TNF-α therapy and interstitial lung disease. Pharmacoepidemiol. Drug Saf. 2013, 22, 394–402. [Google Scholar] [CrossRef] [Green Version]
  182. Campa, M.; Mansouri, B.; Warren, R.; Menter, A. A Review of Biologic Therapies Targeting IL-23 and IL-17 for Use in Moderate-to-Severe Plaque Psoriasis. Dermatol. Ther. 2016, 6, 1–12. [Google Scholar] [CrossRef] [PubMed]
  183. Durham, A.L.; Caramori, G.; Chung, K.F.; Adcock, I.M. Targeted anti-inflammatory therapeutics in asthma and chronic obstructive lung disease. Transl. Res. 2016, 167, 192–203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Choy, D.F.; Hart, K.M.; Borthwick, L.A.; Shikotra, A.; Nagarkar, D.R.; Siddiqui, S.; Jia, G.; Ohri, C.M.; Doran, E.; Vannella, K.M.; et al. TH2 and TH17 inflammatory pathways are reciprocally regulated in asthma. Sci. Transl. Med. 2015, 7, 301ra129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Bhavani, S.; Tsai, C.-L.; Perusich, S.; Hesselbacher, S.; Coxson, H.; Pandit, L.; Corry, D.B.; Kheradmand, F. Clinical and Immunological Factors in Emphysema Progression. Five-Year Prospective Longitudinal Exacerbation Study of Chronic Obstructive Pulmonary Disease (LES-COPD). Am. J. Respir. Crit. Care Med. 2015, 192, 1171–1178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Wilson, M.S.; Madala, S.K.; Ramalingam, T.R.; Gochuico, B.R.; Rosas, I.O.; Cheever, A.W.; Wynn, T.A. Bleomycin and IL-1beta-mediated pulmonary fibrosis is IL-17A dependent. J. Exp. Med. 2010, 207, 535–552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Senoo, S.; Taniguchi, A.; Itano, J.; Oda, N.; Morichika, D.; Fujii, U.; Guo, L.; Sunami, R.; Kanehiro, A.; Tokioka, F.; et al. Essential role of IL-23 in the development of acute exacerbation of pulmonary fibrosis. Am. J. Physiol. Lung Cell. Mol. Physiol. 2021, 321, L925–L940. [Google Scholar] [CrossRef]
  188. Fujii, U.; Miyahara, N.; Taniguchi, A.; Waseda, K.; Morichika, D.; Kurimoto, E.; Koga, H.; Kataoka, M.; Gelfand, E.W.; Cua, D.J.; et al. IL-23 Is Essential for the Development of Elastase-Induced Pulmonary Inflammation and Emphysema. Am. J. Respir. Cell Mol. Biol. 2016, 55, 697–707. [Google Scholar] [CrossRef] [Green Version]
  189. Montani, D.; Günther, S.; Dorfmüller, P.; Perros, F.; Girerd, B.; Garcia, G.; Jaïs, X.; Savale, L.; Artaud-Macari, E.; Price, L.C.; et al. Pulmonary arterial hypertension. Orphanet J. Rare Dis. 2013, 8, 97. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Potential mechanisms for the risk of lung diseases in psoriasis.
Figure 1. Potential mechanisms for the risk of lung diseases in psoriasis.
Ijms 23 01767 g001
Figure 2. Literature search and review methodology.
Figure 2. Literature search and review methodology.
Ijms 23 01767 g002
Figure 3. Literature selection process with subdivisions into selected lung diseases. Abbreviations: COPD, chronic obstructive pulmonary disease; OSA, obstructive sleep apnea; PAH, pulmonary atrial hypertension; ILD, interstitial lung disease.
Figure 3. Literature selection process with subdivisions into selected lung diseases. Abbreviations: COPD, chronic obstructive pulmonary disease; OSA, obstructive sleep apnea; PAH, pulmonary atrial hypertension; ILD, interstitial lung disease.
Ijms 23 01767 g003
Figure 4. Risk factors common to psoriasis and lung diseases. Abbreviations: CTD, connective tissue disorder; COPD, chronic obstructive pulmonary disease; IPF, idiopathic pulmonary fibrosis; PAH, pulmonary atrial hypertension; and OSA, obstructive sleep apnea [43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62].
Figure 4. Risk factors common to psoriasis and lung diseases. Abbreviations: CTD, connective tissue disorder; COPD, chronic obstructive pulmonary disease; IPF, idiopathic pulmonary fibrosis; PAH, pulmonary atrial hypertension; and OSA, obstructive sleep apnea [43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62].
Ijms 23 01767 g004
Figure 5. Immunological similarities between psoriasis and lung diseases. Abbreviations: Th, T helper cell; IL, interleukin [89,90,91,92,93,94].
Figure 5. Immunological similarities between psoriasis and lung diseases. Abbreviations: Th, T helper cell; IL, interleukin [89,90,91,92,93,94].
Ijms 23 01767 g005
Figure 6. Potential abnormalities in the lungs of patients with psoriasis [94,101,102,103,111,112,117,118,120,121].
Figure 6. Potential abnormalities in the lungs of patients with psoriasis [94,101,102,103,111,112,117,118,120,121].
Ijms 23 01767 g006
Table 1. Summary of studies included in the systematic review.
Table 1. Summary of studies included in the systematic review.
ReferencesStudy DesignCases, No. with Lung Disease/Total No.Controls, No. with Lung Disease/Total No.Males, Case/Control, %Patient Ages, Mean +/− SD, yAge of Controls, Mean +/− SD, yPrevalence RateConfounders Controlled for
Asthma
Wang J et al. [12]Meta-analysis1713/66,77234,220/577,415N.A.All ages, N.A.All ages, N.A.OR 1.32 (95% CI, 1.20–1.46)Age, ethnicity
Augustin M et al. [13]Retrospective cohort study160/131327,319/291,868N.A.<18, N.A.<18, N.A.PR 1.34 (95% CI, 1.14–1.59)N.A.
Fang HY et al. [14]Retrospective cohort study420/10,2881153/41,15252.7/52.7≥20, 43.5 ± 17.0≥20, 43.2 ± 17.1HR 1.38 (95% CI, 1.23-1.54)Age, sex, and comorbidities
Hajdarbegovic E et al. [15]Cross-sectional study19/30114/147N.A./44All ages, N.A.All ages, 54 ± 15.6OR 1.60 (95% CI, 0.77–3.32)N.A.
Lønnberg AS et al. [16]Cross-sectional study151/13852,714/31,993N.A.≥20≥20OR 1.32 (95% CI, 1.11–1.57)Sex, age, smoking, body mass index, and chronic obstructive pulmonary disease
Galili E et al. [17]Cross-sectional study345/311270,363/884,653N.A.16–18, N.A.16–18, N.A.OR 1.44 (95% CI, 1.29–1.61)Age, sex, BMI, socio-economic status, country of origin, and number of siblings
Kim SY et al. [18]Retrospective cohort study196/32563,751/164,96344.1/44.1<15<15IR 3.94 (95% CI, 2.16 to 7.17)Age, sex, income, region of residence, and comorbidity
Egeberg A et al. [19]Retrospective cohort study87/658621,638/1,456,385N.A.6–14, N.A.6–14, N.A.IRR 3.85 (95% CI, 2.15–6.90)N.A.
Chronic Obstructive Pulmonary Disease
Ungprasert P et al. [20]Meta-analysisN.A./331,347N.A.N.A.N.A.N.A.OR 1.45 (95% CI, 1.21–1.73)N.A.
Li X et al. [21]Meta-analysis6673/42,15014,368/163,174N.A.N.A.N.A.OR 1.90 (95% CI, 1.36–2.65)N.A.
Wu CY et al. [22]Case-control studyN.A./1127N.A./112746.3/46.3N.A., 53.1N.A., 52.5OR 1.68 (95% CI, 1.02–2.77)None
Chiang YY et al. [23]Retrospective cohort study25/207142/8342N.A.N.A.N.A.HR 2.43 (95% CI, 1.48–3.98)N.A.
Al-Mutairi N et al. [24]Retrospective case-control study98/183574/183552.5/52.5N.A., 52.3 ± 11.9N.A., 52.7 ± 13.5OR 1.46 (95% CI, 1.06–2.01)Drug, smoking status
Obstructive Sleep Apnea
Ger TY et al. [25]Meta-analysisN.A.N.A.N.A.N.A.N.A.OR 2.60 (95% CI, 1.07–6.32)Age, sex, and body mass index
Saçmacı H et al. [26]Cross-sectional study18/604/6050/50N.A., 42.8 ± 13.1N.A., 43.6 ± 13.9OR 6 (95% CI, 1.89–19.04)N.A.
Papadavid E et al. [27]Case-control study24/27229/33079.2/79>18, 50.7 ± 13.6>18, 53 ± 11.8OR 13.31 (95% CI, 1.19–48.93)Age, sex, overweight/obesity, central obesity, comorbidity, and smoking status
Shalom G et al. [28]Case-control study327/12,336369/24,00852.2/50.4>20, 55.6 ± 16.3>20, 54.0 ± 17.1OR 1.27, 95% CI, 1.08–1.49Age, sex, ethnicity, body mass index, and comorbidity
Papadavid E et al. [29]Cross-sectional study19/35N.A.65.7/N.A.N.A., 48.9 ± 13.06N.A.N.A.Age, sex, body mass index, comorbidity, and smoking status
Yang YW et al. [30]Cohort study11/225825/11,25562.6/62.618–59, N.A.18–59, N.A.30 (95% CI = 1.13–4.69)Patients’ monthly incomes, geographic location, urbanization level, and obesity
Pulmonary Atrial Hypertension
Choi YM et al. [31]Retrospective cohort study221/13,936817/69,36048.1/48.1N.A. 57.1 ± 14.86 in mild psoriasis group, 52.4 ± 13.68 in severe psoriasis groupN.A. 57.6 ± 14.84 in mild psoriasis group, 52.9 ± 13.67 in severe psoriasis group1.25 (1.05–1.49) in mild psoriasis group, 1.55 (1.16–2.06) in severe psoriasis groupAge, sex, comorbidity, and medications
Interstitial Lung Disease
Ishikawa G et al. [32]Case series study21/N.A.426/N.A.66.7/N.A.≥20, 66 ± 20≥20, N.A.N.A.Age, sex, comorbidity, body mass index, smoking, and medications
Gupta R et al. [33]Case reportN.A.N.A.N.A.N.A.N.A.N.A.N.A.
Sarcoidosis
Khalid U et al. [34]Cohort study100/70,1259717/5,973,39348.3/49.5≥10, 42.2 ± 18.3 in mild psoriasis group, 41.0 ± 16.6 in severe psoriasis group≥10, 37.0 ± 21.8IR 1.18 (CI, 1.15–1.20) Age, sex, comorbidity, concomitant medications, and socioeconomic status
N.A. = not available/not applicable; OR = odds ratio; PR = prevalence ratio; HR = hazard ratio; IR = incidence ratio; and IRR = incidence rate ratio.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

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. https://doi.org/10.3390/ijms23031767

AMA Style

Mleczko M, Gerkowicz A, Krasowska D. Chronic Inflammation as the Underlying Mechanism of the Development of Lung Diseases in Psoriasis: A Systematic Review. International Journal of Molecular Sciences. 2022; 23(3):1767. https://doi.org/10.3390/ijms23031767

Chicago/Turabian Style

Mleczko, Mateusz, Agnieszka Gerkowicz, and Dorota Krasowska. 2022. "Chronic Inflammation as the Underlying Mechanism of the Development of Lung Diseases in Psoriasis: A Systematic Review" International Journal of Molecular Sciences 23, no. 3: 1767. https://doi.org/10.3390/ijms23031767

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop