Next Article in Journal
Synthesis of Co3O4 Nanoparticles-Decorated Bi12O17Cl2 Hierarchical Microspheres for Enhanced Photocatalytic Degradation of RhB and BPA
Next Article in Special Issue
Reduced Elastin Fibers and Melanocyte Loss in Vitiliginous Skin Are Restored after Repigmentation by Phototherapy and/or Autologous Minigraft Transplantation
Previous Article in Journal
The Epigenetic Role of miR-124 in HIV-1 Tat- and Cocaine-Mediated Microglial Activation
Previous Article in Special Issue
Transforming Growth Factorβ1 Overexpression Is Associated with Insulin Resistance and Rapidly Progressive Kidney Fibrosis under Diabetic Conditions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathological Roles of Pulmonary Cells in Acute Lung Injury: Lessons from Clinical Practice

by
Noriyuki Enomoto
1,2
1
Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
2
Health Administration Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu 431-3192, Japan
Int. J. Mol. Sci. 2022, 23(23), 15027; https://doi.org/10.3390/ijms232315027
Submission received: 24 October 2022 / Revised: 23 November 2022 / Accepted: 26 November 2022 / Published: 30 November 2022
(This article belongs to the Special Issue Cell Death in Acute Organ Injury and Fibrosis)

Abstract

:
Interstitial lung diseases (ILD) are relatively rare and sometimes become life threatening. In particular, rapidly progressive ILD, which frequently presents as acute lung injury (ALI) on lung histopathology, shows poor prognosis if proper and immediate treatments are not initiated. These devastating conditions include acute exacerbation of idiopathic pulmonary fibrosis (AE-IPF), clinically amyopathic dermatomyositis (CADM), epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI)-induced lung injury, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) infection named coronavirus disease 2019 (COVID-19). In this review, clinical information, physical findings, laboratory examinations, and findings on lung high-resolution computed tomography and lung histopathology are presented, focusing on majorly damaged cells in each disease. Furthermore, treatments that should be immediately initiated in clinical practice for each disease are illustrated to save patients with these diseases.

1. Introduction

Interstitial lung disease (ILD) is a relatively rare pathological condition that can induce respiratory insufficiency. In particular, rapidly progressive ILD frequently causes acute respiratory failure and death. Notably, diffuse alveolar damage (DAD) pattern on HRCT or on lung histopathological specimens are tied to a poor prognosis [1,2,3,4]. This rapidly progressive ILD with a poor prognosis includes acute exacerbation of idiopathic pulmonary fibrosis (AE-IPF), clinically amyopathic dermatomyositis (CADM), epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI)-induced lung injury, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) infection, named coronavirus disease 2019 (COVID-19). These devastating diseases frequently cause acute lung injury (ALI), including DAD, although the damaged pulmonary cells, which sometimes become apoptotic, are quite different in each disease. In this review, we focus on the specific diseases that induce severe respiratory failure in clinical practice and the pathological roles of pulmonary cells in ALI.

2. Acute Exacerbation of IPF

Acute exacerbation occurs within approximately one month of the clinical course of chronic progressive ILD, such as IPF [5,6,7]. Three cases of AE-IPF were first reported by Kondoh et al. in 1993 [8]. More AE-IPF occurs in Japanese patients than in Caucasian patients [9]. Therefore, racial and genetic predispositions to AE-IPF should exist in Japanese patients; however, this remains to be elucidated in detail. AE-IPF frequently occurs in elderly patients with end-stage IPF [10,11]. Baseline cardiovascular diseases and higher GAP stage (sex, age, physiology) were significant predictors of AE-IPF [10]. In patients with AE-IPF, lung ground-glass attenuation (GGA) and consolidation opacities appear on chronic fibrotic opacities on high-resolution computed tomography (HRCT) within approximately one month [5,7,12,13]. HRCT and histopathological findings of representative patients with AE-IPF are shown in Figure 1. The appearance of AE leads to poor prognosis in a staircase pattern [14] during the entire long-term course of IPF [15,16]. After the onset of AE, the 90-day mortality reached to 42.9–48.0% [17], and AE is one of the most frequent causes of death and accounts for 30–40% of the causes of deaths [18,19]. AE occurs more frequently in patients with IPF, at the rate of incidence 5.85–14.2%/person-year [15,16,20,21], than in those with other chronic ILD [21,22,23,24]. Patients with connective tissue diseases (CTD) experience AE-ILD at the rate of incidence 1.25-3.3%/person-year [22,23,24], and those with idiopathic non-specific interstitial pneumonia (iNSIP) experience AE-NSIP at the rate of incidence 4.2%/person-year [22]. Therefore, most of the knowledge of AE-ILD came from studies on AE-IPF because of the high frequency of AE-ILD in patients with IPF. With regard to CTD-ILD, AE-ILD most frequently occurs in patients with rheumatoid arthritis (RA) compared to other CTD [11] and has a poor prognosis after the onset of AE, similar to that in AE-IPF [11,24,25]. Furthermore, patients with unclassifiable IIP (UCIIP) also experience AE-UCIIP and show a poor prognosis after the onset of AE, similar to that in AE-IPF [21].
The diagnostic criteria for AE-IPF are as follows: (1) previous or concurrent diagnosis of IPF; (2) worsening of dyspnoea typically <1 month; (3) new bilateral GGA and/or consolidation on CT superimposed on the pattern of usual interstitial pneumonia (UIP); and (4) deterioration not fully explained by cardiac failure or fluid overload [17]. Although these criteria were prepared for AE of IPF, they seem to be applicable for other ILDs instead of IPF/UIP. Furthermore, AE-IPF consists of two subtypes: idiopathic and triggered AE, which include infection, post-operative AEs, drug toxicity, and aspiration [7]. The incidence of AE-IPF is high in winter [26,27]. Therefore, any respiratory infection may have contributed to the increased occurrence of AE-IPF. Torque teno virus was found by microarrays in patients with AE-IPF, and SARS-CoV2 infection was associated with a worse prognosis in those with AE-ILD than in AE-ILD without SARS-CoV2 infection [28]. Furthermore, vaccines against SARS-CoV2 reportedly may have induced AE-IPF [29], although the details remain to be elucidated. AE also occurs after events such as surgical operations [30], including surgical lung biopsy (SLB) [31,32] and bronchoalveolar lavage [33]. Even when a definite diagnosis of AE-IPF is not reached, suspected cases show poor prognosis, similar to definitively diagnosed patients with AE-IPF [27]. Therefore, patients with suspected AE-IPF should be immediately treated with steroids, similar to the treatment for AE-IPF, with concomitant administration of antibiotics for the treatment of causative conditions.
On HRCT, new lung GGA and consolidation opacities appear on chronic fibrotic opacities within approximately one month (Figure 1A,B) [1,5,7]. HRCT findings in AE-IPF are divided into three groups: peripheral, multifocal, and diffuse [12]. Diffuse pattern and large extent of alveolar opacity are significantly related to poor survival [13,34].
In lung histopathology, in addition to collagen deposition in the subpleural alveolar wall consistent with UIP, newly appeared DAD and neutrophil infiltration to the inner normal area are frequently seen in representative cases of AE-IPF (Figure 1C–E) [2,7]. In fact, in post-mortem autopsy specimens, a DAD pattern was observed in 78.8% of AE-IPF cases, while 28.8% exhibited pulmonary haemorrhage, and 17.3% developed thromboembolism [1]. Therefore, various histopathological findings exist in AE-IPF, and cautious diagnostic approaches are required for the histopathological diagnosis of AE-IPF when lung specimens are obtained, although surgical lung biopsy is not a prerequisite for its diagnosis.
As for pathogenesis of AE-IPF, related cells and molecules were summarized in Table 1. The levels of Krebs von den Lungen-6 (KL-6) [2,21], surfactant protein-D (SP-D) [2,21], α-defensin [35], and periostin [36,37] in the blood further increase in AE-IPF compared to those in the chronic phase, which are produced from damaged alveolar epithelial cells, and changes in KL-6 levels [38] or periostin [37] were significant prognostic factors in patients with AE-ILD. In addition, lactate dehydrogenase (LDH) levels increase in AE-IPF [2,21]. Activated neutrophils play an important role in the pathogenesis of AE-ILD/IPF [16,39,40,41], and the high number of neutrophils in bronchoalveolar lavage (BAL) is related to worse prognosis [40]. Furthermore, S100A8 (MRP8) and S100A9 (MRP14), calcium-binding proteins produced released mainly by activated neutrophils, are significant prognostic biomarkers (Figure 1D,E) [2]. Matrix metalloproteinase (MMP)-9, which is produced by activated neutrophils and facilitates the permeability of lung vessels, increases at AE-IPF [39]. In addition, interleukin-8 (IL-8) in BAL, which is related to the migration of neutrophils, also increases in AE-IPF [41]. Activated macrophages also play a role in AE-IPF pathogenesis. C-C motif chemokine ligand (CCL) 18, which is produced by activated macrophages and stimulates macrophages, known as profibrotic-M2 chemokine, increases in BAL in AE-IPF [41,42]. High levels of CCL18 in BAL [41] and higher hemosiderin scores of macrophages [43] are predictive for the development of future AE-ILD. Furthermore, activated macrophages produce ferritin, which is a significant prognostic factor of AE-IPF [44]. In the chronic stage of IPF, single-cell RNA-sequencing revealed an increased number of alveolar macrophages, dendritic cells (DC), and memory T-cells in the fibrotic lung [45]. In collaboration with profibrotic/M2 macrophages, CD4+helper T (Th)-cells such as Th2, Th17, and regulatory T-cell (Tregs) facilitate the profibrotic process [46]. These reports indicate that activated macrophages play a role in the pathogenesis of IPF, not only in the chronic stage, but also in AE-IPF. With regard to gastric aspiration and microorganisms in AE-IPF, higher levels of pepsin are found in BAL fluid in AE-IPF [47], and corisin peptide, which is derived from Staphylococcus and induces apoptosis of lung epithelial cells, is increased in both BAL fluid and serum in AE-IPF [48,49]. As for lung-protective molecules, the functions of heat shock protein (HSP) 70 and thrombomodulin have been reported. HSP70 is a functional TLR4 ligand that promotes endothelial cell survival during lethal oxidant injury [50]. Anti-HSP70 autoantibody was detected in 70% of patients with AE-IPF, while it was detected in 25% of those with IPF in chronic stage and 3% of healthy controls [51]. Thrombomodulin can bind to high-mobility group protein B1 (HMGB1) and prevent HMGB1 from binding to receptors for advanced glycation end-products (RAGE), consequently suppressing inflammation [52,53]. Thrombomodulin decreased [52,53], whereas HMGB1 increased [52,54,55] in AE-IPF. Adipocytes produce more adipokines in AE-IPF, such as adiponectin and leptin, and the adiponectin/leptin ratio is a significant prognostic marker for AE-IPF [56]. Other biomarkers for the prediction of AE-IPF appearance include a decrease of 10% or more in the percent predicted forced vital capacity (%FVC) within 6 months and baseline AaDO2 [57]. Other prognostic biomarkers for patients with AE-IPF include the baseline PaO2/FiO2 (P/F) ratio at AE [16,34], low baseline FVC and diffusion lung capacity for carbon monoxide (%DLCO) [26], C-reactive protein (CRP) [34], delay before initiating therapy [26], ΔP/F ratio 2 days after commencement of treatment for AE, and ΔLDH 2 days after commencement of AE treatment [58]. Recently, a risk-scoring system that predicts 3-month mortality has been reported [34]. This risk scoring system (PCR index) includes the P/F ratio, CRP level, and HRCT pattern, and this system could have well-segregated the prognosis of patients with AE-IPF [34].
As for treatments, AE-IPF may be treated with corticosteroids [59]; however, immunosuppressive therapy is often harmful in the chronic phase of IPF [60]. Steroid pulse therapy is frequently used as the initial therapy for AE-IPF, although evidence for this therapy is not strong enough. The addition of cyclophosphamide [25,61] or recombinant thrombomodulin [62] did not improve the survival of AE-IPF patients [62]. In contrast, a retrospective study showed that the use of antifibrotic agents (pirfenidone or nintedanib) improved the survival of patients with AE-IPF [63]. Furthermore, longitudinal use of nintedanib suppresses AE-IPF [64]. Therefore, antifibrotic agents should be a prerequisite underlying treatment for both acute and chronic stages in patients with IPF. As mentioned previously, activated neutrophils play an important role in the pathogenesis of AE-IPF [16,39,40]. Retrospective studies have reported that long-duration (6 h or more) direct hemoperfusion with a polymyxin B-immobilised fibre column (PMX-DHP) removed activated neutrophils [39] and significantly improved survival in patients with AE-IPF [58,65,66,67]. Furthermore, early commencement of long-duration PMX-DHP was effective in improving the survival of AE-IPF patients compared to later commencement of PMX-DHP [68,69]. PMX-DHP for AE-IPF is promising, but larger prospective studies are needed.

3. Clinically Amyopathic Dermatomyositis-Related ILD

Connective tissue diseases (CTD) sometimes show lung involvement such as ILD, pulmonary hypertension, bronchiolitis, bronchiectasis/bronchiolectasis, and serositis, including pleuritis/pericarditis, depending on the CTD [70]. These lung involvements lead to a wide variety of lung opacities on HRCT and histopathological findings in lung specimens. As for prognosis, particularly the presence of ILD or pulmonary hypertension, is associated with poor survival [71,72,73]. However, the prognosis of patients with CTD-ILD is significantly better than that of IPF patients [74]. CTD-related ILD is frequently seen in patients with systemic sclerosis (SSc) or in those with polymyositis (PM)/dermatomyositis (DM) [70], and ILD accounts for 35% of the causes of death in SSc [71], 48% in PM/DM [73], and 11% in rheumatoid arthritis (RA) [75]. PM/DM-ILD progresses more rapidly [76,77] than SSc-ILD [78] in many cases. Patients with PM/DM-ILD with acute/subacute onset show poorer survival than those with chronic onset [76,79,80]. In addition, DM-ILD shows worse survival than PM-ILD, and clinically amyopathic DM (CADM)-ILD has an even worse prognosis than DM-ILD or PM-ILD [77]. The definition of CADM includes both amyopathic and hypomyopathic DM [81]. Basic and clinical studies on disease-related antibodies have progressed in the field of myositis, and there are two major categories of autoantibodies: myositis-specific antibodies and myositis-associated antibodies [82]. The former includes anti-melanoma differentiation-associated gene (MDA) 5 antibody (previously called anti-CADM 140 antibody) and anti-amynoacyl-tRNA synthetase (ARS) antibodies. These two antibodies are exclusive and closely related to ILD rather than to myositis, although the prognoses differ widely among patients with each antibody. DM-ILD with anti-ARS antibody progresses slowly and responds well to corticosteroids; however, recurrence is not rare [80,83,84]. In contrast, DM-ILD with anti-MDA5 antibody progresses rapidly, is refractory to corticosteroid therapy, and shows poor survival [80,83,84,85,86]. MDA5 is a retinoic acid-inducible gene-I (RIG-I) family of intracellular viral sensors, and the positive rates of anti-MDA5 antibodies have been reported as 14.5–21.5% in DM [87,88] and 25% in PM/DM [80].
In patients with PM/DM and anti-MDA5 antibody, 53.3–82% of them have CADM [80,88,89,90], 94–95% have ILD [88,89], and 71–84% show rapidly progressive ILD [88] or acute/subacute (within 3 months) onset of ILD [80], which has a poor prognosis. Many patients with anti-MDA5 antibody live near the waterfront, and DM-ILD with anti-MDA5 antibody develops predominantly in October-March [90,91]. The exact reason for this is still unknown; however, MDA5 is known to recognise RNA viruses, and the induction of anti-MDA5 antibodies may be related to viral infection.
HRCT findings of DM-ILD with anti-MDA5 antibody are mainly consolidation/GGA and random GGA in lower lung field, and these opacities occupied 83.3% of all cases [92] (Figure 2A,B). Furthermore, 60% of these patients with such opacities on HRCT died despite intensive immunosuppressive treatments [92] (Figure 2C,D). Similarly, the HRCT findings of anti-MDA5 antibody-positive cases are mainly consistent with the unclassifiable pattern of IIPs (66.7%) [80]. As for lung histopathological pattern, five of six cases of DM or CADM had a DAD pattern, and all these patients with DAD died of the progression of ILD [3]. Similarly, seven of nine patients with CADM-ILD with anti-MDA5 antibody had DAD in the lung histopathological specimens [93].
As for pathogenesis of CADM-ILD, related cells and molecules were summarized in Table 1. Activated macrophages play a key role in the pathogenesis of CADM-ILD with anti-MDA5 antibody. Patients with CADM-ILD and anti-MDA5 antibody exhibit activated alveolar macrophages producing ferritin, similar to cells in the bone marrow, liver, and spleen [94]. These findings look similar to macrophage activation syndrome with hyperferritinemic syndrome [95]. In DM-ILD, increased serum ferritin levels negatively correlate with the P/F ratio [96], and patients with higher ferritin levels show a significantly worse survival and serum ferritin level is a significant prognostic factor [96,97]. In addition, in patients with DM-ILD and anti-MDA5 antibody, high macrophage hemophagocytic scores are related to higher ferritin levels [98]. The serum level of CD206, which is preferentially expressed on the surface of alternatively activated (M2) macrophages, was significantly increased in patients with CADM/DM-ILD and anti-MDA5 antibody [97]. M2 macrophages are closely related to tissue repair and fibrosis and the interaction between M2 macrophages and alveolar epithelial cells may be crucial [99]. Similarly, CD163 is also expressed on alveolar macrophages, especially on M2 macrophages [100], and serum levels of soluble CD163 are significantly higher in patients with DM-ILD than in those with PM or without ILD [101]. Furthermore, serum chitotriosidase [102], which macrophages/neutrophils produce, and YKL-40 [103], which is a chitinase family and macrophages/epithelial cells produce, increased in PM/DM-ILD, especially in DM-ILD with anti-MDA5 antibody. As for prognosis related with above mentioned molecules, higher level of serum ferritin [96,97], CD206 [97], CD163 [104], chitotriosidase [102], YKL-40 [103], and hemophagocytic scores [98] are tied to a poor prognosis. Taken together, as indicated above, activated macrophages contribute to the pathophysiology of DM-ILD with anti-MDA5 antibody. The role of monocytes in this process has also been investigated. CCL2 and interferon-induced protein with tetratricopeptide repeats (IFIT) 3 mRNA expression in monocytes and serum CCL2 and interferon (IFN)-β levels are increased in patients with DM-ILD and anti-MDA5 antibody [105]. In addition, serum CCL2 levels are significantly higher in patients with DM-ILD and anti-MDA5 antibody [106]. Both macrophages and monocytes produce CCL2, and the migration of monocytes to the lungs, depending on CCL2, may play a role in the pathogenesis of DM-ILD with anti-MDA5 antibody. In contrast, low circulating lymphocytes and monocytes are found in patients with DM-ILD and anti-MDA5 antibody [107]. In addition, higher serum CCL2 levels [106] and the lower numbers of lymphocytes and monocytes in peripheral blood [107] are associated with a poor survival. The H-ferritin subunit can inhibit lymphoid and myeloid cell proliferation via the T-cell immunoglobulin and mucin domains (TIM) 2 [100,108]. Therefore, H-ferritin produced by macrophages may be related to lower lymphocyte and monocyte counts in blood in patients with DM-ILD and anti-MDA5 antibody. Other immune cells and cytokines, such as neutrophils [109] and IL-8 [109,110], are also associated with a poor prognosis in patients with DM-ILD and anti-MDA5 antibody.
In terms of therapy, when PM/DM-ILD is slowly progressive with chronic onset without anti-MDA5 or anti-ARS antibodies, corticosteroids alone may be sufficient for treatment. However, when anti-MDA5 or anti-ARS antibodies are positive or patients show acute/subacute onset, corticosteroids and calcineurin inhibitors (tacrolimus or cyclosporine A) should be administered [111,112]. Furthermore, patients with acute/subacute onset and anti-MDA5 antibody or patients with rapidly progressive ILD should be treated with corticosteroids, calcineurin inhibitors, and intravenous cyclophosphamide (IVCY) [113,114]. This triple combination therapy significantly improves survival compared to historical controls [115]. However, triple combination therapy significantly increases the incidence of serious infections [115], with an odds ratio of 5.51 [116]. Therefore, overtreatment should be avoided in DM-ILD patients without rapidly progressive ILD or anti-MDA5 antibody [113,114]. Moreover, if DM-ILD is refractory to the above-mentioned treatments, intravenous immunoglobulin (IVIG) should be considered [117,118]. Finally, when DM or CADM-ILD progress despite appropriate treatment, this type of ILD is recently called progressive fibrosing (PF)-ILD [119] or progressive pulmonary fibrosis (PPF) [59]. If progressive cases meet the criteria for PF-ILD or PPF, patients with DM or CADM-ILD should be treated with an anti-fibrotic agent (nintedanib) [119] following the above-mentioned immunosuppressive treatments.

4. EGFR-TKI-Induced Lung Injury

The number of patients with drug-induced lung injury (DLI) has increased with the development of new medicines. Especially in Japan, the number of DLI has strikingly increased since 2000 [120], and the representative causative drugs are EGFR-TKIs against non-small cell lung cancer (NSCLC), including gefitinib [4] and erlotinib [121], and antirheumatic drugs including leflunomide [122]. In the 2000s, severe DLI and death due to gefitinib were frequently reported [4] which was soon a serious public concern in Japan [120]. Gefitinib-induced lung injury was observed in 5.8% of Japanese patients with NSCLC, and the mortality rate was 38.6% [120]. The incidence of gefitinib-induced lung injury was higher than that of chemotherapy within the first four weeks (odds ratio, 3.2 [123]. Furthermore, erlotinib-induced lung injury was found in 4.3% of Japanese NSCLC patients, and the mortality rate was as high as 35.7% [121]. Risk factors for the development of EGFR-TKI-induced lung injury include the presence of comorbid ILD, smoking history, male sex, and performance status of two or more [120]. In particular, the presence of preceding ILD and smoking history are common risk factors for EGFR-TKIs [120] and leflunomide [122]. Moreover, the presence of preceding ILD was related to increased mortality in EGFR-TKI-induced lung injuries [123]. Therefore, screening for preceding ILD using chest HRCT before initiating EGFR-TKI therapy is important to prevent DLI and DLI-induced death in clinical practice. Japanese patients are predisposed to EGFR-TKI-induced lung injury compared to people of non-Japanese origin [124], and the odds ratio of all pneumonitis was 5.04 and that of grade 5 pneumonitis (death) was 4.55 [124]. Additionally, the frequency of EGFR-TKI-induced lung injury was not significantly different between Asian and non-Asian populations but was significantly higher in Japanese than in non-Japanese Asians (odds ratio 12.7) [125].
The HRCT patterns of DLI consist of five patterns: DAD, NSIP, organising pneumonia (OP), hypersensitivity pneumonitis (HP), and acute eosinophilic pneumonia (AEP) [126]. A CT image of the gefitinib-induced lung injury is shown in Figure 3A [4]. This case shows extensive diffuse GGA consistent with a DAD pattern. In this way, when patients show a DAD pattern on HRCT, they have the lowest P/F ratio, and the steroid cumulative dose is highest among these five patterns [126]. Patients with a DAD pattern on HRCT show significantly higher serum KL-6 levels, and those with more opacities on HRCT have higher serum KL-6 levels [127]. BAL in DAD includes many neutrophils [120]. In terms of EGFR-TKI-induced lung injury, patients frequently show DAD patterns on both HRCT (Figure 3A) and histopathology (Figure 3B), and many patients with a DAD pattern have a poor prognosis [4,120], with a mortality rate of approximately 40% [121]. Therefore, careful attention should be paid when patients with EGFR-TKI-induced lung injury exhibit a DAD pattern on HRCT. Moreover, infections such as pneumocystis pneumonia should be ruled out, especially when patients are immunocompromised or treated with corticosteroids, immunosuppressants, or biological agents [128].
In the pathogenesis of EGFR-TKI-induced lung injury, related cells and molecules were summarized in Table 1. Gefitinib induces lung inflammation via the production of IL-1β, and the release of HMGB1 from macrophages further leads to the death of macrophages [129,130]. This inflammation and subsequent programmed cell death is known as pyroptosis [129]. Gefitinib causes mitochondrial damage and mitochondrial reactive oxygen species (ROS) production. Mitochondrial ROS then activate the NLR family pyrin domain-containing protein (NLRP) 3 inflammasome and facilitate the production of IL-1β [129,130]. Furthermore, mitochondrial ROS induce DNA damage and facilitate the release of HMGB1, which can initiate a positive feedback loop of NLRP3 inflammasome signalling, leading to excessive inflammation [129,130]. In contrast, HSP70 protects lungs against pulmonary fibrosis, and administration of gefitinib suppresses the expression of HSP70 in the lungs and facilitates pulmonary fibrosis in a rodent model [131]. As written in the part of AE-ILD, HSP70 is a functionally TLR4 ligand during lethal oxidant injury that promotes endothelial cell survival [50]. Anti-HSP70 autoantibodies were detected in 70% of patients with AE-IPF compared to 3% of healthy controls [51]. Administration of geranylgeranylacetone, an inducer of HSP70, decreases lung inflammation and apoptosis of lung cells and further suppresses bleomycin-induced pulmonary fibrosis [132]. Furthermore, administration of geranylgeranylacetone suppresses gefitinib-induced exacerbation of pulmonary fibrosis [131]. Therefore, given these basic experimental results, the administration of NLR3-inhibitor, neutralising antibody of IL-1β, papaverine as an inhibitor of HMGB1 [130], and geranylgeranylacetone may be promising for the future treatment of EGFR-TKI-induced lung injury.

5. COVID-19 and ILD

SARS-CoV2 is an enveloped positive-sense single-stranded RNA virus. The SARS-CoV2 outbreak occurred in Wuhan at the end of 2019 and spread worldwide thereafter. This infection by SARS-CoV2 was named COVID-19 and was declared to be a pandemic in March 2020 by the World Health Organization (WHO). As of October 2022, more than 613 million people have been infected with SARS-CoV2 and more than 6.5 million deaths have occurred worldwide (information on WHO COVID-19 dashboard: https://covid19.who.int). COVID-19 is now the most frequent infectious disease in the world far exceeding the prevalence of tuberculosis. The excess mortality rate exceeded 300 deaths per 100,000 people in 21 countries from January 2020 to December 2021 [133]. Notably, the mortality risk was higher in the Delta variant pandemic period than in the Omicron variant period [134]. Although the clinical presentation of COVID-19 is highly variable, some patients experience hypoxaemia without discomfort, which is called “silent hypoxemia” [135]. Thereafter, in some cases, COVID-19 causes severe acute respiratory distress syndrome (ARDS), particularly in patients with risk factors such as older age, male sex, cardiovascular disease, chronic respiratory disease, diabetes, obesity, and hypertension [136,137]. Patients with pre-existing ILD, such as IPF, RA-ILD, or SSc-ILD, show worse survival than those without pre-existing ILD [138]. Furthermore, patients with COVID-19-related AE-ILD have a worse prognosis than those without COVID-19 [28]. In patients without pre-existing ILD, those with COVID-19 ARDS have a higher body mass index and longer duration of mechanical ventilation than those with non-COVID-19 ARDS; however, 60-day mortality is similar [139]. In addition, thromboses such as pulmonary embolism and venous thromboembolism, which may be related to hypoxia, occur more frequently in COVID-19 especially in severe COVID-19 cases than in other infectious pneumonia or in mild to moderate COVID-19 cases, with an incidence of 9.5–30% [140,141,142]. Microthrombi in capillaries are found in fatal COVID-19-associated lung injuries [143]. On HRCT, multiple GGA, crazy paving patterns, and consolidation are seen in the peripheral or peribronchiolar lung area in many cases with COVID-19 [142,144] (Figure 4). These findings on HRCT change over time by phases (Figure 4A, early phase; B, 10 days later; and C, another 7 days later) [142], and the more diffuse extent of the findings on CT is associated with higher severity of COVID-19 [142]. Moreover, pulmonary vessel enlargement is observed in or adjacent to GGA and consolidation [142,145] (Figure 4D). Hypoxic pulmonary vasoconstriction (HPV) may be impaired due to endothelial damage caused by SARS-CoV-2 infection and may be related to pulmonary vessel enlargement on HRCT [137,146]. In addition, as mentioned above, thromboses such as pulmonary embolism are seen in severe COVID-19 patients (Figure 4E,F) [142]. In autopsy lung specimens of patients with fatal COVID-19, DAD patterns with excessive thrombosis and injury to alveolar epithelial cells/endothelial cells have been observed [137,147,148].
SARS-CoV-2 invades human cells, including alveolar epithelial cells, vascular endothelial cells, and lymphocytes, via the angiotensin-converting enzyme (ACE) 2 receptor [149] and CD147 [150]. In the pathogenesis of COVID-19 and ILD, related cells and molecules were summarized in Table 1. Cellular senescence and mitochondrial dysfunction play important roles [149]. Mitochondrial dysfunction and apoptosis are observed in lymphocytes, especially T cells, which are important for protection from SARS-CoV-2 [151], and are related to lymphocytopenia in patients with COVID-19 [150,152,153]. Mitochondrial ROS and subsequent activation of the NLRP3 inflammasome appear to be related to severe respiratory failure in coronavirus infection [154,155]. Furthermore, abnormal mitochondrial ultrastructure and increased expression of inhibitory checkpoints, such as programmed death-1 (PD-1) and its ligand (PD-L1), are found in monocytes in patients with COVID-19 [156]; and non-classical monocytes are further decreased in those with severe COVID-19 [157]. In line with these reports, T cells also show higher levels of the exhausted marker PD-1 and reduced expression of CXCR6 [158], which is important for the localisation of resident memory T cells, and the number of CD4+ and CD8+ T cells is reduced [159,160]. Moreover, the reduction in CD4+ and CD8+ T cells is negatively correlated with survival in patients with COVID-19 [160]. Additionally, cytokine production and reactivation of SARS-CoV-2 specific CD8+ T cells are inhibited in severe COVID-19 cases [161]. Similarly, apoptosis in plasmacytoid dendritic cells (DC) [162] and the decreased number of plasmacytoid DC and myeloid DC are found in patients with COVID-19 [163], which may be related to the impaired protective function by type 1 IFN. Additionally, neutrophils accumulate in the lungs, and calprotectin (S100A8/S100A9), which is a calcium-binding protein mainly produced from activated neutrophils, promotes inflammation [164] and increases in blood in patients with severe COVID-19 [157]. Neutrophils and neutrophil extracellular traps (NETs) are abundantly present in seriously damaged COVID-19 lung tissue [143], similar to ALI in influenza pneumonitis [165]. Regarding lung cell death, SARS-CoV-2 proteins and induced cytokines lead to PANoptosis consisting of apoptosis, pyroptosis, and necroptosis in the same cell population, which is a unique inflammatory programmed cell death [155,166]. SARS-CoV-2-induced synergistic effect of tumor necrosis factor (TNF)-α and IFN-γ causes PANoptosis [166]. Additionally, SARS-CoV-2-induced activation of caspase-8 causes apoptosis [167]. In this pathway, phosphorylation of receptor-interacting protein kinase-3 (RIPK3) and mixed lineage kinase domain-like (MLKL) also induces necroptosis [167] and facilitates inflammation via IL-1β, which may be related to COVID-19-induced ARDS. In fact, serum level of RIPK3 is significantly higher in severe COVID-19 cases than in mild cases [168]. Furthermore, the open reading frame (ORF) 3a, a SARS-CoV-2 accessory viroprotein, induces apoptosis via activation of caspase-8 [169] or enhances pyroptosis of infected cells [155].
In terms of prophylaxis, newly developed messenger RNA (mRNA) vaccines not only significantly reduce the number of infected patients [170,171], but also decrease the severity of COVID-19, although the efficacy rates to prevent infection decreased in the Omicron variant pandemic period in 2022 [134,172]. As for treatments, early use of antiviral medicines and late use of corticosteroids seem to be beneficial [173]. As of October 2022, in the viral replication phase, starting nirmatrelvir/ritonavir [173,174] or Molnupiravir [175] within 3–5 days of symptom onset reduce the risk of hospitalisation or death. Baricitinib [176], a Janus kinase (JAK) inhibitor, or Baricitinib with remdesivir [177] reduces the time to recovery or 28-day mortality, however, remdesivir alone does not reduce 28-day mortality [178]. Monoclonal antibodies target spike proteins and decrease symptom duration and mortality against the Delta variant but not against the Omicron variant, except for sotrovimab [173]. In the inflammatory phase, 6 mg/day of dexamethasone for 10 days decreases 28-day mortality in hospitalised patients with severe COVID-19 [179], although a higher dose of methylprednisolone may also be effective. Tocilizumab [180], an IL-6 receptor antagonist, and prophylactic anticoagulation [173] are also effective in critically ill patients with COVID-19.

6. Conclusions

In this review, four diseases, AE of IPF, CADM, EGFR-TKI-induced lung injury, and COVID-19, which lead to rapid progressive ILD and respiratory failure, are reviewed. All these conditions have poor prognoses and frequently show ALI, including DAD, on HRCT and lung histopathological specimens. However, pathological cells in the lungs, which are mainly impacted and sometimes become apoptotic, are quite different in each disease. These cells include alveolar epithelial cells, pulmonary vascular endothelial cells, alveolar macrophages, lymphocytes, and neutrophils, which play important roles in the pathogenesis of various diseases. Therefore, attending doctors should carefully monitor patients with these diseases to check their clinical information, physical findings, laboratory examinations, and lung HRCT findings, and immediately initiate proper treatments to save these patients. Further novel medical developments that prevent and treat these diseases will be desired in the future.

Funding

This study did not receive any funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This work was supported by the Study Group on Diffuse Lung Disease and the Scientific Research/Research on Intractable Diseases of the Ministry of Health, Labour, and Welfare of Japan.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Oda, K.; Ishimoto, H.; Yamada, S.; Kushima, H.; Ishii, H.; Imanaga, T.; Harada, T.; Ishimatsu, Y.; Matsumoto, N.; Naito, K.; et al. Autopsy analyses in acute exacerbation of idiopathic pulmonary fibrosis. Respir. Res. 2014, 15, 109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Tanaka, K.; Enomoto, N.; Hozumi, H.; Isayama, T.; Naoi, H.; Aono, Y.; Katsumata, M.; Yasui, H.; Karayama, M.; Suzuki, Y.; et al. Serum S100A8 and S100A9 as prognostic biomarkers in acute exacerbation of idiopathic pulmonary fibrosis. Respir. Investig. 2021, 59, 827–836. [Google Scholar] [CrossRef] [PubMed]
  3. Matsuki, Y.; Yamashita, H.; Takahashi, Y.; Kano, T.; Shimizu, A.; Itoh, K.; Kaneko, H.; Mimori, A. Diffuse alveolar damage in patients with dermatomyositis: A six-case series. Mod. Rheumatol. 2012, 22, 243–248. [Google Scholar] [CrossRef] [PubMed]
  4. Inoue, A.; Saijo, Y.; Maemondo, M.; Gomi, K.; Tokue, Y.; Kimura, Y.; Ebina, M.; Kikuchi, T.; Moriya, T.; Nukiwa, T. Severe acute interstitial pneumonia and gefitinib. Lancet 2003, 361, 137–139. [Google Scholar] [CrossRef] [PubMed]
  5. Collard, H.R.; Moore, B.B.; Flaherty, K.R.; Brown, K.K.; Kaner, R.J.; King, T.E., Jr.; Lasky, J.A.; Loyd, J.E.; Noth, I.; Olman, M.A.; et al. Acute exacerbations of idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 2007, 176, 636–643. [Google Scholar] [CrossRef] [Green Version]
  6. Raghu, G.; Collard, H.R.; Egan, J.J.; Martinez, F.J.; Behr, J.; Brown, K.K.; Colby, T.V.; Cordier, J.F.; Flaherty, K.R.; Lasky, J.A.; et al. An official ATS/ERS/JRS/ALAT statement: Idiopathic pulmonary fibrosis: Evidence-based guidelines for diagnosis and management. Am. J. Respir. Crit. Care Med. 2011, 183, 788–824. [Google Scholar] [CrossRef] [Green Version]
  7. Collard, H.R.; Ryerson, C.J.; Corte, T.J.; Jenkins, G.; Kondoh, Y.; Lederer, D.J.; Lee, J.S.; Maher, T.M.; Wells, A.U.; Antoniou, K.M.; et al. Acute Exacerbation of Idiopathic Pulmonary Fibrosis. An International Working Group Report. Am. J. Respir. Crit. Care Med. 2016, 194, 265–275. [Google Scholar] [CrossRef]
  8. Kondoh, Y.; Taniguchi, H.; Kawabata, Y.; Yokoi, T.; Suzuki, K.; Takagi, K. Acute exacerbation in idiopathic pulmonary fibrosis. Analysis of clinical and pathologic findings in three cases. Chest 1993, 103, 1808–1812. [Google Scholar] [CrossRef]
  9. Azuma, A.; Hagiwara, K.; Kudoh, S. Basis of acute exacerbation of idiopathic pulmonary fibrosis in Japanese patients. Am. J. Respir. Crit. Care Med. 2008, 177, 1397–1398. [Google Scholar] [CrossRef]
  10. Kakugawa, T.; Sakamoto, N.; Sato, S.; Yura, H.; Harada, T.; Nakashima, S.; Hara, A.; Oda, K.; Ishimoto, H.; Yatera, K.; et al. Risk factors for an acute exacerbation of idiopathic pulmonary fibrosis. Respir. Res. 2016, 17, 79. [Google Scholar] [CrossRef]
  11. Enomoto, N.; Oyama, Y.; Enomoto, Y.; Yasui, H.; Karayama, M.; Kono, M.; Hozumi, H.; Suzuki, Y.; Furuhashi, K.; Fujisawa, T.; et al. Differences in clinical features of acute exacerbation between connective tissue disease-associated interstitial pneumonia and idiopathic pulmonary fibrosis. Chron. Respir. Dis. 2019, 16, 1479972318809476. [Google Scholar] [CrossRef] [PubMed]
  12. Akira, M.; Hamada, H.; Sakatani, M.; Kobayashi, C.; Nishioka, M.; Yamamoto, S. CT findings during phase of accelerated deterioration in patients with idiopathic pulmonary fibrosis. AJR Am. J. Roentgenol. 1997, 168, 79–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Akira, M.; Kozuka, T.; Yamamoto, S.; Sakatani, M. Computed tomography findings in acute exacerbation of idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 2008, 178, 372–378. [Google Scholar] [CrossRef] [PubMed]
  14. Kim, D.S. Acute exacerbations in patients with idiopathic pulmonary fibrosis. Respir. Res. 2013, 14, 86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Kondoh, Y.; Taniguchi, H.; Katsuta, T.; Kataoka, K.; Kimura, T.; Nishiyama, O.; Sakamoto, K.; Johkoh, T.; Nishimura, M.; Ono, K.; et al. Risk factors of acute exacerbation of idiopathic pulmonary fibrosis. Sarcoidosis Vasc. Diffuse Lung Dis. 2010, 27, 103–110. [Google Scholar]
  16. Song, J.W.; Hong, S.B.; Lim, C.M.; Koh, Y.; Kim, D.S. Acute exacerbation of idiopathic pulmonary fibrosis: Incidence, risk factors and outcome. Eur. Respir. J. 2011, 37, 356–363. [Google Scholar] [CrossRef]
  17. Collard, H.R.; Richeldi, L.; Kim, D.S.; Taniguchi, H.; Tschoepe, I.; Luisetti, M.; Roman, J.; Tino, G.; Schlenker-Herceg, R.; Hallmann, C.; et al. Acute exacerbations in the INPULSIS trials of nintedanib in idiopathic pulmonary fibrosis. Eur. Respir. J. 2017, 49, 1601339. [Google Scholar] [CrossRef] [Green Version]
  18. Ley, B.; Collard, H.R.; King, T.E., Jr. Clinical course and prediction of survival in idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 2011, 183, 431–440. [Google Scholar] [CrossRef]
  19. Natsuizaka, M.; Chiba, H.; Kuronuma, K.; Otsuka, M.; Kudo, K.; Mori, M.; Bando, M.; Sugiyama, Y.; Takahashi, H. Epidemiologic survey of Japanese patients with idiopathic pulmonary fibrosis and investigation of ethnic differences. Am. J. Respir. Crit. Care Med. 2014, 190, 773–779. [Google Scholar] [CrossRef]
  20. Kim, D.S.; Park, J.H.; Park, B.K.; Lee, J.S.; Nicholson, A.G.; Colby, T. Acute exacerbation of idiopathic pulmonary fibrosis: Frequency and clinical features. Eur. Respir. J. 2006, 27, 143–150. [Google Scholar] [CrossRef] [Green Version]
  21. Enomoto, N.; Naoi, H.; Aono, Y.; Katsumata, M.; Horiike, Y.; Yasui, H.; Karayama, M.; Hozumi, H.; Suzuki, Y.; Furuhashi, K.; et al. Acute exacerbation of unclassifiable idiopathic interstitial pneumonia: Comparison with idiopathic pulmonary fibrosis. Ther. Adv. Respir. Dis. 2020, 14, 1753466620935774. [Google Scholar] [CrossRef] [PubMed]
  22. Park, I.N.; Kim, D.S.; Shim, T.S.; Lim, C.M.; Lee, S.D.; Koh, Y.; Kim, W.S.; Kim, W.D.; Jang, S.J.; Colby, T.V. Acute exacerbation of interstitial pneumonia other than idiopathic pulmonary fibrosis. Chest 2007, 132, 214–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Suda, T.; Kaida, Y.; Nakamura, Y.; Enomoto, N.; Fujisawa, T.; Imokawa, S.; Hashizume, H.; Naito, T.; Hashimoto, D.; Takehara, Y.; et al. Acute exacerbation of interstitial pneumonia associated with collagen vascular diseases. Respir. Med. 2009, 103, 846–853. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Enomoto, N.; Naoi, H.; Mochizuka, Y.; Isayama, T.; Tanaka, Y.; Fukada, A.; Aono, Y.; Katsumata, M.; Yasui, H.; Mori, K.; et al. Frequency, proportion of PF-ILD, and prognostic factors in patients with acute exacerbation of ILD related to systemic autoimmune diseases. BMC Pulm. Med. 2022, 22, 387. [Google Scholar] [CrossRef]
  25. Hozumi, H.; Kono, M.; Hasegawa, H.; Kato, S.; Inoue, Y.; Suzuki, Y.; Karayama, M.; Furuhashi, K.; Enomoto, N.; Fujisawa, T.; et al. Acute exacerbation of rheumatoid arthritis-associated interstitial lung disease: Mortality and its prediction model. Respir. Res. 2022, 23, 57. [Google Scholar] [CrossRef]
  26. Simon-Blancal, V.; Freynet, O.; Nunes, H.; Bouvry, D.; Naggara, N.; Brillet, P.Y.; Denis, D.; Cohen, Y.; Vincent, F.; Valeyre, D.; et al. Acute exacerbation of idiopathic pulmonary fibrosis: Outcome and prognostic factors. Respiration 2012, 83, 28–35. [Google Scholar] [CrossRef]
  27. Collard, H.R.; Yow, E.; Richeldi, L.; Anstrom, K.J.; Glazer, C. Suspected acute exacerbation of idiopathic pulmonary fibrosis as an outcome measure in clinical trials. Respir. Res. 2013, 14, 73. [Google Scholar] [CrossRef] [Green Version]
  28. Kondoh, Y.; Kataoka, K.; Ando, M.; Awaya, Y.; Ichikado, K.; Kataoka, M.; Komase, Y.; Mineshita, M.; Ohno, Y.; Okamoto, H.; et al. COVID-19 and acute exacerbation of interstitial lung disease. Respir. Investig. 2021, 59, 675–678. [Google Scholar] [CrossRef]
  29. Sgalla, G.; Magri, T.; Lerede, M.; Comes, A.; Richeldi, L. COVID-19 Vaccine in Patients with Exacerbation of Idiopathic Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2022, 206, 219–221. [Google Scholar] [CrossRef]
  30. Sato, T.; Teramukai, S.; Kondo, H.; Watanabe, A.; Ebina, M.; Kishi, K.; Fujii, Y.; Mitsudomi, T.; Yoshimura, M.; Maniwa, T.; et al. Impact and predictors of acute exacerbation of interstitial lung diseases after pulmonary resection for lung cancer. J. Thorac. Cardiovasc. Surg. 2014, 147, 1604–1611.e3. [Google Scholar] [CrossRef] [Green Version]
  31. Kondoh, Y.; Taniguchi, H.; Kitaichi, M.; Yokoi, T.; Johkoh, T.; Oishi, T.; Kimura, T.; Nishiyama, O.; Kato, K.; du Bois, R.M. Acute exacerbation of interstitial pneumonia following surgical lung biopsy. Respir. Med. 2006, 100, 1753–1759. [Google Scholar] [CrossRef]
  32. Kaarteenaho, R. The current position of surgical lung biopsy in the diagnosis of idiopathic pulmonary fibrosis. Respir. Res. 2013, 14, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Sakamoto, K.; Taniguchi, H.; Kondoh, Y.; Wakai, K.; Kimura, T.; Kataoka, K.; Hashimoto, N.; Nishiyama, O.; Hasegawa, Y. Acute exacerbation of IPF following diagnostic bronchoalveolar lavage procedures. Respir. Med. 2012, 106, 436–442. [Google Scholar] [CrossRef] [Green Version]
  34. Sakamoto, S.; Shimizu, H.; Isshiki, T.; Nakamura, Y.; Usui, Y.; Kurosaki, A.; Isobe, K.; Takai, Y.; Homma, S. New risk scoring system for predicting 3-month mortality after acute exacerbation of idiopathic pulmonary fibrosis. Sci. Rep. 2022, 12, 1134. [Google Scholar] [CrossRef]
  35. Konishi, K.; Gibson, K.F.; Lindell, K.O.; Richards, T.J.; Zhang, Y.; Dhir, R.; Bisceglia, M.; Gilbert, S.; Yousem, S.A.; Song, J.W.; et al. Gene expression profiles of acute exacerbations of idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 2009, 180, 167–175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Izuhara, K.; Conway, S.J.; Moore, B.B.; Matsumoto, H.; Holweg, C.T.; Matthews, J.G.; Arron, J.R. Roles of Periostin in Respiratory Disorders. Am. J. Respir. Crit. Care Med. 2016, 193, 949–956. [Google Scholar] [CrossRef] [Green Version]
  37. Shimizu, H.; Sakamoto, S.; Okamoto, M.; Isshiki, T.; Ono, J.; Shimizu, S.; Hoshino, T.; Izuhara, K.; Homma, S. Association of serum monomeric periostin level with outcomes of acute exacerbation of idiopathic pulmonary fibrosis and fibrosing nonspecific interstitial pneumonia. Ann. Transl. Med. 2021, 9, 739. [Google Scholar] [CrossRef] [PubMed]
  38. Choi, M.G.; Choi, S.M.; Lee, J.H.; Yoon, J.K.; Song, J.W. Changes in blood Krebs von den Lungen-6 predict the mortality of patients with acute exacerbation of interstitial lung disease. Sci. Rep. 2022, 12, 4916. [Google Scholar] [CrossRef]
  39. Abe, S.; Seo, Y.; Hayashi, H.; Matsuda, K.; Usuki, J.; Azuma, A.; Kudoh, S.; Gemma, A. Neutrophil adsorption by polymyxin B-immobilized fiber column for acute exacerbation in patients with interstitial pneumonia: A pilot study. Blood Purif. 2010, 29, 321–326. [Google Scholar] [CrossRef]
  40. Kono, M.; Miyashita, K.; Hirama, R.; Oshima, Y.; Takeda, K.; Mochizuka, Y.; Tsutsumi, A.; Miwa, H.; Miki, Y.; Hashimoto, D.; et al. Prognostic significance of bronchoalveolar lavage cellular analysis in patients with acute exacerbation of interstitial lung disease. Respir. Med. 2021, 186, 106534. [Google Scholar] [CrossRef]
  41. Schupp, J.C.; Binder, H.; Jager, B.; Cillis, G.; Zissel, G.; Muller-Quernheim, J.; Prasse, A. Macrophage activation in acute exacerbation of idiopathic pulmonary fibrosis. PLoS ONE 2015, 10, e0116775. [Google Scholar] [CrossRef]
  42. Prasse, A.; Probst, C.; Bargagli, E.; Zissel, G.; Toews, G.B.; Flaherty, K.R.; Olschewski, M.; Rottoli, P.; Muller-Quernheim, J. Serum CC-chemokine ligand 18 concentration predicts outcome in idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 2009, 179, 717–723. [Google Scholar] [CrossRef] [PubMed]
  43. Arai, T.; Kagawa, T.; Sasaki, Y.; Sugawara, R.; Sugimoto, C.; Tachibana, K.; Fujita, Y.; Hayashi, S.; Inoue, Y. Hemosiderin-Laden Macrophages in Bronchoalveolar Lavage: Predictive Role for Acute Exacerbation of Idiopathic Interstitial Pneumonias. Can. Respir. J. 2021, 2021, 4595019. [Google Scholar] [CrossRef] [PubMed]
  44. Enomoto, N.; Oyama, Y.; Enomoto, Y.; Mikamo, M.; Karayama, M.; Hozumi, H.; Suzuki, Y.; Kono, M.; Furuhashi, K.; Fujisawa, T.; et al. Prognostic evaluation of serum ferritin in acute exacerbation of idiopathic pulmonary fibrosis. Clin. Respir. J. 2018, 12, 2378–2389. [Google Scholar] [CrossRef] [PubMed]
  45. Serezani, A.P.M.; Pascoalino, B.D.; Bazzano, J.M.R.; Vowell, K.N.; Tanjore, H.; Taylor, C.J.; Calvi, C.L.; McCall, A.S.; Bacchetta, M.D.; Shaver, C.M.; et al. Multiplatform Single-Cell Analysis Identifies Immune Cell Types Enhanced in Pulmonary Fibrosis. Am. J. Respir. Cell Mol. Biol. 2022, 67, 50–60. [Google Scholar] [CrossRef] [PubMed]
  46. Shenderov, K.; Collins, S.L.; Powell, J.D.; Horton, M.R. Immune dysregulation as a driver of idiopathic pulmonary fibrosis. J. Clin. Investig. 2021, 131, e143226. [Google Scholar] [CrossRef]
  47. Lee, J.S.; Song, J.W.; Wolters, P.J.; Elicker, B.M.; King, T.E., Jr.; Kim, D.S.; Collard, H.R. Bronchoalveolar lavage pepsin in acute exacerbation of idiopathic pulmonary fibrosis. Eur. Respir. J. 2012, 39, 352–358. [Google Scholar] [CrossRef]
  48. D’Alessandro-Gabazza, C.N.; Yasuma, T.; Kobayashi, T.; Toda, M.; Abdel-Hamid, A.M.; Fujimoto, H.; Hataji, O.; Nakahara, H.; Takeshita, A.; Nishihama, K.; et al. Inhibition of lung microbiota-derived proapoptotic peptides ameliorates acute exacerbation of pulmonary fibrosis. Nat. Commun. 2022, 13, 1558. [Google Scholar] [CrossRef]
  49. D’Alessandro-Gabazza, C.N.; Kobayashi, T.; Yasuma, T.; Toda, M.; Kim, H.; Fujimoto, H.; Hataji, O.; Takeshita, A.; Nishihama, K.; Okano, T.; et al. A Staphylococcus pro-apoptotic peptide induces acute exacerbation of pulmonary fibrosis. Nat. Commun. 2020, 11, 1539. [Google Scholar] [CrossRef] [Green Version]
  50. Zhang, Y.; Zhang, X.; Shan, P.; Hunt, C.R.; Pandita, T.K.; Lee, P.J. A protective Hsp70-TLR4 pathway in lethal oxidant lung injury. J. Immunol. 2013, 191, 1393–1403. [Google Scholar] [CrossRef] [Green Version]
  51. Kahloon, R.A.; Xue, J.; Bhargava, A.; Csizmadia, E.; Otterbein, L.; Kass, D.J.; Bon, J.; Soejima, M.; Levesque, M.C.; Lindell, K.O.; et al. Patients with idiopathic pulmonary fibrosis with antibodies to heat shock protein 70 have poor prognoses. Am. J. Respir. Crit. Care Med. 2013, 187, 768–775. [Google Scholar] [CrossRef] [PubMed]
  52. Ebina, M.; Taniguchi, H.; Miyasho, T.; Yamada, S.; Shibata, N.; Ohta, H.; Hisata, S.; Ohkouchi, S.; Tamada, T.; Nishimura, H.; et al. Gradual increase of high mobility group protein b1 in the lungs after the onset of acute exacerbation of idiopathic pulmonary fibrosis. Pulm. Med. 2011, 2011, 916486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Collard, H.R.; Calfee, C.S.; Wolters, P.J.; Song, J.W.; Hong, S.B.; Brady, S.; Ishizaka, A.; Jones, K.D.; King, T.E., Jr.; Matthay, M.A.; et al. Plasma biomarker profiles in acute exacerbation of idiopathic pulmonary fibrosis. Am. J. Physiol. Lung Cell Mol. Physiol. 2010, 299, L3–L7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Abe, S.; Hayashi, H.; Seo, Y.; Matsuda, K.; Kamio, K.; Saito, Y.; Usuki, J.; Azuma, A.; Kudo, S.; Gemma, A. Reduction in serum high mobility group box-1 level by polymyxin B-immobilized fiber column in patients with idiopathic pulmonary fibrosis with acute exacerbation. Blood Purif. 2011, 32, 310–316. [Google Scholar] [CrossRef]
  55. Yamaguchi, K.; Iwamoto, H.; Sakamoto, S.; Horimasu, Y.; Masuda, T.; Miyamoto, S.; Nakashima, T.; Ohshimo, S.; Fujitaka, K.; Hamada, H.; et al. Serum high-mobility group box 1 is associated with the onset and severity of acute exacerbation of idiopathic pulmonary fibrosis. Respirology 2020, 25, 275–280. [Google Scholar] [CrossRef]
  56. Enomoto, N.; Oyama, Y.; Yasui, H.; Karayama, M.; Hozumi, H.; Suzuki, Y.; Kono, M.; Furuhashi, K.; Fujisawa, T.; Inui, N.; et al. Analysis of serum adiponectin and leptin in patients with acute exacerbation of idiopathic pulmonary fibrosis. Sci. Rep. 2019, 9, 10484. [Google Scholar] [CrossRef] [Green Version]
  57. Kondoh, Y.; Taniguchi, H.; Ebina, M.; Azuma, A.; Ogura, T.; Taguchi, Y.; Suga, M.; Takahashi, H.; Nakata, K.; Sugiyama, Y.; et al. Risk factors for acute exacerbation of idiopathic pulmonary fibrosis--Extended analysis of pirfenidone trial in Japan. Respir. Investig. 2015, 53, 271–278. [Google Scholar] [CrossRef]
  58. Enomoto, N.; Mikamo, M.; Oyama, Y.; Kono, M.; Hashimoto, D.; Fujisawa, T.; Inui, N.; Nakamura, Y.; Yasuda, H.; Kato, A.; et al. Treatment of acute exacerbation of idiopathic pulmonary fibrosis with direct hemoperfusion using a polymyxin B-immobilized fiber column improves survival. BMC Pulm. Med. 2015, 15, 15. [Google Scholar] [CrossRef] [Green Version]
  59. Raghu, G.; Remy-Jardin, M.; Richeldi, L.; Thomson, C.C.; Inoue, Y.; Johkoh, T.; Kreuter, M.; Lynch, D.A.; Maher, T.M.; Martinez, F.J.; et al. Idiopathic Pulmonary Fibrosis (an Update) and Progressive Pulmonary Fibrosis in Adults: An Official ATS/ERS/JRS/ALAT Clinical Practice Guideline. Am. J. Respir. Crit. Care Med. 2022, 205, e18–e47. [Google Scholar] [CrossRef]
  60. Idiopathic Pulmonary Fibrosis Clinical Research, N.; Raghu, G.; Anstrom, K.J.; King, T.E., Jr.; Lasky, J.A.; Martinez, F.J. Prednisone, azathioprine, and N-acetylcysteine for pulmonary fibrosis. N. Engl. J. Med. 2012, 366, 1968–1977. [Google Scholar] [CrossRef]
  61. Naccache, J.M.; Jouneau, S.; Didier, M.; Borie, R.; Cachanado, M.; Bourdin, A.; Reynaud-Gaubert, M.; Bonniaud, P.; Israel-Biet, D.; Prevot, G.; et al. Cyclophosphamide added to glucocorticoids in acute exacerbation of idiopathic pulmonary fibrosis (EXAFIP): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir. Med. 2022, 10, 26–34. [Google Scholar] [CrossRef] [PubMed]
  62. Kondoh, Y.; Azuma, A.; Inoue, Y.; Ogura, T.; Sakamoto, S.; Tsushima, K.; Johkoh, T.; Fujimoto, K.; Ichikado, K.; Matsuzawa, Y.; et al. Thrombomodulin Alfa for Acute Exacerbation of Idiopathic Pulmonary Fibrosis. A Randomized, Double-Blind Placebo-controlled Trial. Am. J. Respir. Crit. Care Med. 2020, 201, 1110–1119. [Google Scholar] [CrossRef] [PubMed]
  63. Kang, J.; Han, M.; Song, J.W. Antifibrotic treatment improves clinical outcomes in patients with idiopathic pulmonary fibrosis: A propensity score matching analysis. Sci. Rep. 2020, 10, 15620. [Google Scholar] [CrossRef] [PubMed]
  64. Richeldi, L.; Cottin, V.; du Bois, R.M.; Selman, M.; Kimura, T.; Bailes, Z.; Schlenker-Herceg, R.; Stowasser, S.; Brown, K.K. Nintedanib in patients with idiopathic pulmonary fibrosis: Combined evidence from the TOMORROW and INPULSIS((R)) trials. Respir. Med. 2016, 113, 74–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Abe, S.; Azuma, A.; Mukae, H.; Ogura, T.; Taniguchi, H.; Bando, M.; Sugiyama, Y. Polymyxin B-immobilized fiber column (PMX) treatment for idiopathic pulmonary fibrosis with acute exacerbation: A multicenter retrospective analysis. Intern. Med. 2012, 51, 1487–1491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Enomoto, N.; Suda, T.; Uto, T.; Kato, M.; Kaida, Y.; Ozawa, Y.; Miyazaki, H.; Kuroishi, S.; Hashimoto, D.; Naito, T.; et al. Possible therapeutic effect of direct haemoperfusion with a polymyxin B immobilized fibre column (PMX-DHP) on pulmonary oxygenation in acute exacerbations of interstitial pneumonia. Respirology 2008, 13, 452–460. [Google Scholar] [CrossRef] [PubMed]
  67. Kono, M.; Suda, T.; Enomoto, N.; Nakamura, Y.; Kaida, Y.; Hashimoto, D.; Inui, N.; Mizuguchi, T.; Kato, A.; Shirai, T.; et al. Evaluation of different perfusion durations in direct hemoperfusion with polymyxin B-immobilized fiber column therapy for acute exacerbation of interstitial pneumonias. Blood Purif. 2011, 32, 75–81. [Google Scholar] [CrossRef]
  68. Oishi, K.; Aoe, K.; Mimura, Y.; Murata, Y.; Sakamoto, K.; Koutoku, W.; Matsumoto, T.; Ueoka, H.; Yano, M. Survival from an Acute Exacerbation of Idiopathic Pulmonary Fibrosis with or without Direct Hemoperfusion with a Polymyxin B-immobilized Fiber Column: A Retrospective Analysis. Intern. Med. 2016, 55, 3551–3559. [Google Scholar] [CrossRef] [Green Version]
  69. Furusawa, H.; Sugiura, M.; Mitaka, C.; Inase, N. Direct hemoperfusion with polymyxin B-immobilized fibre treatment for acute exacerbation of interstitial pneumonia. Respirology 2017, 22, 1357–1362. [Google Scholar] [CrossRef]
  70. Fischer, A.; du Bois, R. Interstitial lung disease in connective tissue disorders. Lancet 2012, 380, 689–698. [Google Scholar] [CrossRef]
  71. Tyndall, A.J.; Bannert, B.; Vonk, M.; Airo, P.; Cozzi, F.; Carreira, P.E.; Bancel, D.F.; Allanore, Y.; Muller-Ladner, U.; Distler, O.; et al. Causes and risk factors for death in systemic sclerosis: A study from the EULAR Scleroderma Trials and Research (EUSTAR) database. Ann. Rheum. Dis. 2010, 69, 1809–1815. [Google Scholar] [CrossRef] [PubMed]
  72. Hoffmann-Vold, A.M.; Fretheim, H.; Halse, A.K.; Seip, M.; Bitter, H.; Wallenius, M.; Garen, T.; Salberg, A.; Brunborg, C.; Midtvedt, O.; et al. Tracking Impact of Interstitial Lung Disease in Systemic Sclerosis in a Complete Nationwide Cohort. Am. J. Respir. Crit. Care Med. 2019, 200, 1258–1266. [Google Scholar] [CrossRef] [PubMed]
  73. Yamasaki, Y.; Yamada, H.; Ohkubo, M.; Yamasaki, M.; Azuma, K.; Ogawa, H.; Mizushima, M.; Ozaki, S. Longterm survival and associated risk factors in patients with adult-onset idiopathic inflammatory myopathies and amyopathic dermatomyositis: Experience in a single institute in Japan. J. Rheumatol. 2011, 38, 1636–1643. [Google Scholar] [CrossRef] [PubMed]
  74. Park, J.H.; Kim, D.S.; Park, I.N.; Jang, S.J.; Kitaichi, M.; Nicholson, A.G.; Colby, T.V. Prognosis of fibrotic interstitial pneumonia: Idiopathic versus collagen vascular disease-related subtypes. Am. J. Respir. Crit. Care Med. 2007, 175, 705–711. [Google Scholar] [CrossRef]
  75. Nakajima, A.; Inoue, E.; Tanaka, E.; Singh, G.; Sato, E.; Hoshi, D.; Shidara, K.; Hara, M.; Momohara, S.; Taniguchi, A.; et al. Mortality and cause of death in Japanese patients with rheumatoid arthritis based on a large observational cohort, IORRA. Scand. J. Rheumatol. 2010, 39, 360–367. [Google Scholar] [CrossRef]
  76. Suda, T.; Fujisawa, T.; Enomoto, N.; Nakamura, Y.; Inui, N.; Naito, T.; Hashimoto, D.; Sato, J.; Toyoshima, M.; Hashizume, H.; et al. Interstitial lung diseases associated with amyopathic dermatomyositis. Eur. Respir. J. 2006, 28, 1005–1012. [Google Scholar] [CrossRef]
  77. Fujisawa, T.; Hozumi, H.; Kono, M.; Enomoto, N.; Hashimoto, D.; Nakamura, Y.; Inui, N.; Yokomura, K.; Koshimizu, N.; Toyoshima, M.; et al. Prognostic factors for myositis-associated interstitial lung disease. PLoS ONE 2014, 9, e98824. [Google Scholar] [CrossRef]
  78. Distler, O.; Highland, K.B.; Gahlemann, M.; Azuma, A.; Fischer, A.; Mayes, M.D.; Raghu, G.; Sauter, W.; Girard, M.; Alves, M.; et al. Nintedanib for Systemic Sclerosis-Associated Interstitial Lung Disease. N. Engl. J. Med. 2019, 380, 2518–2528. [Google Scholar] [CrossRef] [PubMed]
  79. Fujisawa, T.; Suda, T.; Nakamura, Y.; Enomoto, N.; Ide, K.; Toyoshima, M.; Uchiyama, H.; Tamura, R.; Ida, M.; Yagi, T.; et al. Differences in clinical features and prognosis of interstitial lung diseases between polymyositis and dermatomyositis. J. Rheumatol. 2005, 32, 58–64. [Google Scholar]
  80. Hozumi, H.; Fujisawa, T.; Nakashima, R.; Johkoh, T.; Sumikawa, H.; Murakami, A.; Enomoto, N.; Inui, N.; Nakamura, Y.; Hosono, Y.; et al. Comprehensive assessment of myositis-specific autoantibodies in polymyositis/dermatomyositis-associated interstitial lung disease. Respir. Med. 2016, 121, 91–99. [Google Scholar] [CrossRef] [Green Version]
  81. Sontheimer, R.D. Would a new name hasten the acceptance of amyopathic dermatomyositis (dermatomyositis sine myositis) as a distinctive subset within the idiopathic inflammatory dermatomyopathies spectrum of clinical illness? J. Am. Acad. Dermatol. 2002, 46, 626–636. [Google Scholar] [CrossRef] [PubMed]
  82. Ghirardello, A.; Borella, E.; Beggio, M.; Franceschini, F.; Fredi, M.; Doria, A. Myositis autoantibodies and clinical phenotypes. Autoimmun. Highlights 2014, 5, 69–75. [Google Scholar] [CrossRef] [PubMed]
  83. Hozumi, H.; Enomoto, N.; Kono, M.; Fujisawa, T.; Inui, N.; Nakamura, Y.; Sumikawa, H.; Johkoh, T.; Nakashima, R.; Imura, Y.; et al. Prognostic significance of anti-aminoacyl-tRNA synthetase antibodies in polymyositis/dermatomyositis-associated interstitial lung disease: A retrospective case control study. PLoS ONE 2015, 10, e0120313. [Google Scholar] [CrossRef] [PubMed]
  84. Chen, F.; Li, S.; Wang, T.; Shi, J.; Wang, G. Clinical Heterogeneity of Interstitial Lung Disease in Polymyositis and Dermatomyositis Patients With or Without Specific Autoantibodies. Am. J. Med. Sci. 2018, 355, 48–53. [Google Scholar] [CrossRef] [PubMed]
  85. Nakashima, R.; Imura, Y.; Kobayashi, S.; Yukawa, N.; Yoshifuji, H.; Nojima, T.; Kawabata, D.; Ohmura, K.; Usui, T.; Fujii, T.; et al. The RIG-I-like receptor IFIH1/MDA5 is a dermatomyositis-specific autoantigen identified by the anti-CADM-140 antibody. Rheumatology 2010, 49, 433–440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Tanizawa, K.; Handa, T.; Nakashima, R.; Kubo, T.; Hosono, Y.; Aihara, K.; Ikezoe, K.; Watanabe, K.; Taguchi, Y.; Hatta, K.; et al. The prognostic value of HRCT in myositis-associated interstitial lung disease. Respir. Med. 2013, 107, 745–752. [Google Scholar] [CrossRef] [Green Version]
  87. Labrador-Horrillo, M.; Martinez, M.A.; Selva-O’Callaghan, A.; Trallero-Araguas, E.; Balada, E.; Vilardell-Tarres, M.; Juarez, C. Anti-MDA5 antibodies in a large Mediterranean population of adults with dermatomyositis. J. Immunol. Res. 2014, 2014, 290797. [Google Scholar] [CrossRef] [Green Version]
  88. Koga, T.; Fujikawa, K.; Horai, Y.; Okada, A.; Kawashiri, S.Y.; Iwamoto, N.; Suzuki, T.; Nakashima, Y.; Tamai, M.; Arima, K.; et al. The diagnostic utility of anti-melanoma differentiation-associated gene 5 antibody testing for predicting the prognosis of Japanese patients with DM. Rheumatology 2012, 51, 1278–1284. [Google Scholar] [CrossRef] [Green Version]
  89. Hoshino, K.; Muro, Y.; Sugiura, K.; Tomita, Y.; Nakashima, R.; Mimori, T. Anti-MDA5 and anti-TIF1-gamma antibodies have clinical significance for patients with dermatomyositis. Rheumatology 2010, 49, 1726–1733. [Google Scholar] [CrossRef] [Green Version]
  90. Nishina, N.; Sato, S.; Masui, K.; Gono, T.; Kuwana, M. Seasonal and residential clustering at disease onset of anti-MDA5-associated interstitial lung disease. RMD Open 2020, 6, e001202. [Google Scholar] [CrossRef]
  91. Muro, Y.; Sugiura, K.; Hoshino, K.; Akiyama, M.; Tamakoshi, K. Epidemiologic study of clinically amyopathic dermatomyositis and anti-melanoma differentiation-associated gene 5 antibodies in central Japan. Arthritis Res. Ther. 2011, 13, R214. [Google Scholar] [CrossRef] [Green Version]
  92. Tanizawa, K.; Handa, T.; Nakashima, R.; Kubo, T.; Hosono, Y.; Watanabe, K.; Aihara, K.; Oga, T.; Chin, K.; Nagai, S.; et al. HRCT features of interstitial lung disease in dermatomyositis with anti-CADM-140 antibody. Respir. Med. 2011, 105, 1380–1387. [Google Scholar] [CrossRef] [PubMed]
  93. Chino, H.; Sekine, A.; Baba, T.; Iwasawa, T.; Okudela, K.; Takemura, T.; Itoh, H.; Sato, S.; Suzuki, Y.; Ogura, T. Radiological and Pathological Correlation in Anti-MDA5 Antibody-positive Interstitial Lung Disease: Rapidly Progressive Perilobular Opacities and Diffuse Alveolar Damage. Intern. Med. 2016, 55, 2241–2246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Gono, T.; Miyake, K.; Kawaguchi, Y.; Kaneko, H.; Shinozaki, M.; Yamanaka, H. Hyperferritinaemia and macrophage activation in a patient with interstitial lung disease with clinically amyopathic DM. Rheumatology 2012, 51, 1336–1338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Rosario, C.; Zandman-Goddard, G.; Meyron-Holtz, E.G.; D’Cruz, D.P.; Shoenfeld, Y. The hyperferritinemic syndrome: Macrophage activation syndrome, Still’s disease, septic shock and catastrophic antiphospholipid syndrome. BMC Med. 2013, 11, 185. [Google Scholar] [CrossRef] [Green Version]
  96. Gono, T.; Kawaguchi, Y.; Satoh, T.; Kuwana, M.; Katsumata, Y.; Takagi, K.; Masuda, I.; Tochimoto, A.; Baba, S.; Okamoto, Y.; et al. Clinical manifestation and prognostic factor in anti-melanoma differentiation-associated gene 5 antibody-associated interstitial lung disease as a complication of dermatomyositis. Rheumatology 2010, 49, 1713–1719. [Google Scholar] [CrossRef] [Green Version]
  97. Horiike, Y.; Suzuki, Y.; Fujisawa, T.; Yasui, H.; Karayama, M.; Hozumi, H.; Furuhashi, K.; Enomoto, N.; Nakamura, Y.; Inui, N.; et al. Successful classification of macrophage-mannose receptor CD206 in severity of anti-MDA5 antibody positive dermatomyositis associated ILD. Rheumatology 2019, 58, 2143–2152. [Google Scholar] [CrossRef]
  98. Zhao, S.; Ma, X.; Zhang, X.; Jin, Z.; Hu, W.; Hua, B.; Wang, H.; Feng, X.; Sun, L.; Chen, Z. Clinical significance of HScore and MS score comparison in the prognostic evaluation of anti-MDA5-positive patients with dermatomyositis and interstitial lung disease. Mod. Rheumatol. 2022, 32, 373–379. [Google Scholar] [CrossRef]
  99. Aggarwal, N.R.; King, L.S.; D’Alessio, F.R. Diverse macrophage populations mediate acute lung inflammation and resolution. Am. J. Physiol. Lung Cell Mol. Physiol. 2014, 306, L709–L725. [Google Scholar] [CrossRef]
  100. Colafrancesco, S.; Priori, R.; Alessandri, C.; Astorri, E.; Perricone, C.; Blank, M.; Agmon-Levin, N.; Shoenfeld, Y.; Valesini, G. The hyperferritinemic syndromes and CD163: A marker of macrophage activation. Isr. Med. Assoc. J. 2014, 16, 662–663. [Google Scholar]
  101. Peng, Q.L.; Zhang, Y.L.; Shu, X.M.; Yang, H.B.; Zhang, L.; Chen, F.; Lu, X.; Wang, G.C. Elevated Serum Levels of Soluble CD163 in Polymyositis and Dermatomyositis: Associated with Macrophage Infiltration in Muscle Tissue. J. Rheumatol. 2015, 42, 979–987. [Google Scholar] [CrossRef] [PubMed]
  102. Fujisawa, T.; Hozumi, H.; Yasui, H.; Suzuki, Y.; Karayama, M.; Furuhashi, K.; Enomoto, N.; Nakamura, Y.; Inui, N.; Suda, T. Clinical Significance of Serum Chitotriosidase Level in Anti-MDA5 Antibody-positive Dermatomyositis-associated Interstitial Lung Disease. J. Rheumatol. 2019, 46, 935–942. [Google Scholar] [CrossRef] [PubMed]
  103. Hozumi, H.; Fujisawa, T.; Enomoto, N.; Nakashima, R.; Enomoto, Y.; Suzuki, Y.; Kono, M.; Karayama, M.; Furuhashi, K.; Murakami, A.; et al. Clinical Utility of YKL-40 in Polymyositis/dermatomyositis-associated Interstitial Lung Disease. J. Rheumatol. 2017, 44, 1394–1401. [Google Scholar] [CrossRef] [PubMed]
  104. Enomoto, Y.; Suzuki, Y.; Hozumi, H.; Mori, K.; Kono, M.; Karayama, M.; Furuhashi, K.; Fujisawa, T.; Enomoto, N.; Nakamura, Y.; et al. Clinical significance of soluble CD163 in polymyositis-related or dermatomyositis-related interstitial lung disease. Arthritis Res. Ther. 2017, 19, 9. [Google Scholar] [CrossRef] [Green Version]
  105. Gono, T.; Okazaki, Y.; Kuwana, M. Antiviral proinflammatory phenotype of monocytes in anti-MDA5 antibody-associated interstitial lung disease. Rheumatology 2022, 61, 806–814. [Google Scholar] [CrossRef]
  106. Oda, K.; Kotani, T.; Takeuchi, T.; Ishida, T.; Shoda, T.; Isoda, K.; Yoshida, S.; Nishimura, Y.; Makino, S. Chemokine profiles of interstitial pneumonia in patients with dermatomyositis: A case control study. Sci. Rep. 2017, 7, 1635. [Google Scholar] [CrossRef] [Green Version]
  107. Lv, X.; Jin, Y.; Zhang, D.; Li, Y.; Fu, Y.; Wang, S.; Ye, Y.; Wu, W.; Ye, S.; Yan, B.; et al. Low Circulating Monocytes Is in Parallel with Lymphopenia Which Predicts Poor Outcome in Anti-melanoma Differentiation-Associated Gene 5 Antibody-Positive Dermatomyositis-Associated Interstitial Lung Disease. Front. Med. 2021, 8, 808875. [Google Scholar] [CrossRef]
  108. Chen, T.T.; Li, L.; Chung, D.H.; Allen, C.D.; Torti, S.V.; Torti, F.M.; Cyster, J.G.; Chen, C.Y.; Brodsky, F.M.; Niemi, E.C.; et al. TIM-2 is expressed on B cells and in liver and kidney and is a receptor for H-ferritin endocytosis. J. Exp. Med. 2005, 202, 955–965. [Google Scholar] [CrossRef]
  109. Shirakashi, M.; Nakashima, R.; Tsuji, H.; Tanizawa, K.; Handa, T.; Hosono, Y.; Akizuki, S.; Murakami, K.; Hashimoto, M.; Yoshifuji, H.; et al. Efficacy of plasma exchange in anti-MDA5-positive dermatomyositis with interstitial lung disease under combined immunosuppressive treatment. Rheumatology 2020, 59, 3284–3292. [Google Scholar] [CrossRef]
  110. Gono, T.; Kaneko, H.; Kawaguchi, Y.; Hanaoka, M.; Kataoka, S.; Kuwana, M.; Takagi, K.; Ichida, H.; Katsumata, Y.; Ota, Y.; et al. Cytokine profiles in polymyositis and dermatomyositis complicated by rapidly progressive or chronic interstitial lung disease. Rheumatology 2014, 53, 2196–2203. [Google Scholar] [CrossRef] [Green Version]
  111. Takada, K.; Katada, Y.; Ito, S.; Hayashi, T.; Kishi, J.; Itoh, K.; Yamashita, H.; Hirakata, M.; Kawahata, K.; Kawakami, A.; et al. Impact of adding tacrolimus to initial treatment of interstitial pneumonitis in polymyositis/dermatomyositis: A single-arm clinical trial. Rheumatology 2020, 59, 1084–1093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Fujisawa, T.; Hozumi, H.; Kamiya, Y.; Kaida, Y.; Akamatsu, T.; Kusagaya, H.; Satake, Y.; Mori, K.; Mikamo, M.; Matsuda, H.; et al. Prednisolone and tacrolimus versus prednisolone and cyclosporin A to treat polymyositis/dermatomyositis-associated ILD: A randomized, open-label trial. Respirology 2021, 26, 370–377. [Google Scholar] [CrossRef] [PubMed]
  113. Fujisawa, T. Management of Myositis-Associated Interstitial Lung Disease. Medicina 2021, 57, 347. [Google Scholar] [CrossRef]
  114. Kondoh, Y.; Makino, S.; Ogura, T.; Suda, T.; Tomioka, H.; Amano, H.; Anraku, M.; Enomoto, N.; Fujii, T.; Fujisawa, T.; et al. 2020 guide for the diagnosis and treatment of interstitial lung disease associated with connective tissue disease. Respir. Investig. 2021, 59, 709–740. [Google Scholar] [CrossRef] [PubMed]
  115. Tsuji, H.; Nakashima, R.; Hosono, Y.; Imura, Y.; Yagita, M.; Yoshifuji, H.; Hirata, S.; Nojima, T.; Sugiyama, E.; Hatta, K.; et al. Multicenter Prospective Study of the Efficacy and Safety of Combined Immunosuppressive Therapy with High-Dose Glucocorticoid, Tacrolimus, and Cyclophosphamide in Interstitial Lung Diseases Accompanied by Anti-Melanoma Differentiation-Associated Gene 5-Positive Dermatomyositis. Arthritis Rheumatol. 2020, 72, 488–498. [Google Scholar] [PubMed]
  116. Sugiyama, Y.; Yoshimi, R.; Tamura, M.; Takeno, M.; Kunishita, Y.; Kishimoto, D.; Yoshioka, Y.; Kobayashi, K.; Takase-Minegishi, K.; Watanabe, T.; et al. The predictive prognostic factors for polymyositis/dermatomyositis-associated interstitial lung disease. Arthritis Res. Ther. 2018, 20, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Suzuki, Y.; Hayakawa, H.; Miwa, S.; Shirai, M.; Fujii, M.; Gemma, H.; Suda, T.; Chida, K. Intravenous immunoglobulin therapy for refractory interstitial lung disease associated with polymyositis/dermatomyositis. Lung 2009, 187, 201–206. [Google Scholar] [CrossRef] [PubMed]
  118. Wang, L.M.; Yang, Q.H.; Zhang, L.; Liu, S.Y.; Zhang, P.P.; Zhang, X.; Liu, X.J.; Han, L.S.; Li, T.F. Intravenous immunoglobulin for interstitial lung diseases of anti-melanoma differentiation-associated gene 5-positive dermatomyositis. Rheumatology 2022, 61, 3704–3710. [Google Scholar] [CrossRef]
  119. Flaherty, K.R.; Wells, A.U.; Cottin, V.; Devaraj, A.; Walsh, S.L.F.; Inoue, Y.; Richeldi, L.; Kolb, M.; Tetzlaff, K.; Stowasser, S.; et al. Nintedanib in Progressive Fibrosing Interstitial Lung Diseases. N. Engl. J. Med. 2019, 381, 1718–1727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Kubo, K.; Azuma, A.; Kanazawa, M.; Kameda, H.; Kusumoto, M.; Genma, A.; Saijo, Y.; Sakai, F.; Sugiyama, Y.; Tatsumi, K.; et al. Consensus statement for the diagnosis and treatment of drug-induced lung injuries. Respir. Investig. 2013, 51, 260–277. [Google Scholar] [CrossRef] [Green Version]
  121. Gemma, A.; Kudoh, S.; Ando, M.; Ohe, Y.; Nakagawa, K.; Johkoh, T.; Yamazaki, N.; Arakawa, H.; Inoue, Y.; Ebina, M.; et al. Final safety and efficacy of erlotinib in the phase 4 POLARSTAR surveillance study of 10 708 Japanese patients with non-small-cell lung cancer. Cancer Sci. 2014, 105, 1584–1590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Sawada, T.; Inokuma, S.; Sato, T.; Otsuka, T.; Saeki, Y.; Takeuchi, T.; Matsuda, T.; Takemura, T.; Sagawa, A.; Study Committee for Leflunomide-induced Lung Injury, Japan College of Rheumatology. Leflunomide-induced interstitial lung disease: Prevalence and risk factors in Japanese patients with rheumatoid arthritis. Rheumatology 2009, 48, 1069–1072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Kudoh, S.; Kato, H.; Nishiwaki, Y.; Fukuoka, M.; Nakata, K.; Ichinose, Y.; Tsuboi, M.; Yokota, S.; Nakagawa, K.; Suga, M.; et al. Interstitial lung disease in Japanese patients with lung cancer: A cohort and nested case-control study. Am. J. Respir. Crit. Care Med. 2008, 177, 1348–1357. [Google Scholar] [CrossRef] [PubMed]
  124. Suh, C.H.; Park, H.S.; Kim, K.W.; Pyo, J.; Hatabu, H.; Nishino, M. Pneumonitis in advanced non-small-cell lung cancer patients treated with EGFR tyrosine kinase inhibitor: Meta-analysis of 153 cohorts with 15,713 patients: Meta-analysis of incidence and risk factors of EGFR-TKI pneumonitis in NSCLC. Lung Cancer 2018, 123, 60–69. [Google Scholar] [CrossRef] [PubMed]
  125. Takeda, M.; Okamoto, I.; Nakagawa, K. Pooled safety analysis of EGFR-TKI treatment for EGFR mutation-positive non-small cell lung cancer. Lung Cancer 2015, 88, 74–79. [Google Scholar] [CrossRef] [PubMed]
  126. Takatani, K.; Miyazaki, E.; Nureki, S.; Ando, M.; Ueno, T.; Okubo, T.; Takenaka, R.; Hiroshige, S.; Kumamoto, T. High-resolution computed tomography patterns and immunopathogenetic findings in drug-induced pneumonitis. Respir. Med. 2008, 102, 892–898. [Google Scholar] [CrossRef] [Green Version]
  127. Ohnishi, H.; Yokoyama, A.; Yasuhara, Y.; Watanabe, A.; Naka, T.; Hamada, H.; Abe, M.; Nishimura, K.; Higaki, J.; Ikezoe, J.; et al. Circulating KL-6 levels in patients with drug induced pneumonitis. Thorax 2003, 58, 872–875. [Google Scholar] [CrossRef] [Green Version]
  128. Kameda, H.; Tokuda, H.; Sakai, F.; Johkoh, T.; Mori, S.; Yoshida, Y.; Takayanagi, N.; Taki, H.; Hasegawa, Y.; Hatta, K.; et al. Clinical and radiological features of acute-onset diffuse interstitial lung diseases in patients with rheumatoid arthritis receiving treatment with biological agents: Importance of Pneumocystis pneumonia in Japan revealed by a multicenter study. Intern. Med. 2011, 50, 305–313. [Google Scholar] [CrossRef] [Green Version]
  129. Noguchi, T.; Sekiguchi, Y.; Kudoh, Y.; Naganuma, R.; Kagi, T.; Nishidate, A.; Maeda, K.; Ishii, C.; Toyama, T.; Hirata, Y.; et al. Gefitinib initiates sterile inflammation by promoting IL-1beta and HMGB1 release via two distinct mechanisms. Cell Death Dis. 2021, 12, 49. [Google Scholar] [CrossRef]
  130. Kagi, T.; Noguchi, T.; Matsuzawa, A. Mechanisms of gefitinib-induced interstitial pneumonitis: Why and how the TKI perturbs innate immune systems? Oncotarget 2021, 12, 1321–1322. [Google Scholar] [CrossRef]
  131. Namba, T.; Tanaka, K.; Hoshino, T.; Azuma, A.; Mizushima, T. Suppression of expression of heat shock protein 70 by gefitinib and its contribution to pulmonary fibrosis. PLoS ONE 2011, 6, e27296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Fujibayashi, T.; Hashimoto, N.; Jijiwa, M.; Hasegawa, Y.; Kojima, T.; Ishiguro, N. Protective effect of geranylgeranylacetone, an inducer of heat shock protein 70, against drug-induced lung injury/fibrosis in an animal model. BMC Pulm. Med. 2009, 9, 45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. COVID-19 Excess Mortality Collaborators. Estimating excess mortality due to the COVID-19 pandemic: A systematic analysis of COVID-19-related mortality, 2020–2021. Lancet 2022, 399, 1513–1536. [Google Scholar]
  134. Adjei, S.; Hong, K.; Molinari, N.M.; Bull-Otterson, L.; Ajani, U.A.; Gundlapalli, A.V.; Harris, A.M.; Hsu, J.; Kadri, S.S.; Starnes, J.; et al. Mortality Risk Among Patients Hospitalized Primarily for COVID-19 during the Omicron and Delta Variant Pandemic Periods-United States, April 2020–June 2022. MMWR Morb. Mortal. Wkly. Rep. 2022, 71, 1182–1189. [Google Scholar] [CrossRef]
  135. Simonson, T.S.; Baker, T.L.; Banzett, R.B.; Bishop, T.; Dempsey, J.A.; Feldman, J.L.; Guyenet, P.G.; Hodson, E.J.; Mitchell, G.S.; Moya, E.A.; et al. Silent hypoxaemia in COVID-19 patients. J. Physiol. 2021, 599, 1057–1065. [Google Scholar] [CrossRef]
  136. Terada, M.; Ohtsu, H.; Saito, S.; Hayakawa, K.; Tsuzuki, S.; Asai, Y.; Matsunaga, N.; Kutsuna, S.; Sugiura, W.; Ohmagari, N. Risk factors for severity on admission and the disease progression during hospitalisation in a large cohort of patients with COVID-19 in Japan. BMJ Open 2021, 11, e047007. [Google Scholar] [CrossRef]
  137. Swenson, K.E.; Swenson, E.R. Pathophysiology of Acute Respiratory Distress Syndrome and COVID-19 Lung Injury. Crit. Care Clin. 2021, 37, 749–776. [Google Scholar] [CrossRef]
  138. Zhao, J.; Metra, B.; George, G.; Roman, J.; Mallon, J.; Sundaram, B.; Li, M.; Summer, R. Mortality among Patients with COVID-19 and Different Interstitial Lung Disease Subtypes: A Multicenter Cohort Study. Ann. Am. Thorac. Soc. 2022, 19, 1435–1437. [Google Scholar] [CrossRef]
  139. Bain, W.; Yang, H.; Shah, F.A.; Suber, T.; Drohan, C.; Al-Yousif, N.; DeSensi, R.S.; Bensen, N.; Schaefer, C.; Rosborough, B.R.; et al. COVID-19 versus Non-COVID-19 Acute Respiratory Distress Syndrome: Comparison of Demographics, Physiologic Parameters, Inflammatory Biomarkers, and Clinical Outcomes. Ann. Am. Thorac. Soc. 2021, 18, 1202–1210. [Google Scholar] [CrossRef]
  140. Nishimoto, Y.; Yachi, S.; Takeyama, M.; Tsujino, I.; Nakamura, J.; Yamamoto, N.; Nakata, H.; Ikeda, S.; Umetsu, M.; Aikawa, S.; et al. The current status of thrombosis and anticoagulation therapy in patients with COVID-19 in Japan: From the CLOT-COVID study. J. Cardiol. 2022, 80, 285–291. [Google Scholar] [CrossRef]
  141. Su, H.; Yang, M.; Wan, C.; Yi, L.X.; Tang, F.; Zhu, H.Y.; Yi, F.; Yang, H.C.; Fogo, A.B.; Nie, X.; et al. Renal histopathological analysis of 26 postmortem findings of patients with COVID-19 in China. Kidney Int. 2020, 98, 219–227. [Google Scholar] [CrossRef] [PubMed]
  142. Larici, A.R.; Cicchetti, G.; Marano, R.; Bonomo, L.; Storto, M.L. COVID-19 pneumonia: Current evidence of chest imaging features, evolution and prognosis. Chin. J. Acad. Radiol. 2021, 4, 229–240. [Google Scholar] [CrossRef] [PubMed]
  143. Obermayer, A.; Jakob, L.M.; Haslbauer, J.D.; Matter, M.S.; Tzankov, A.; Stoiber, W. Neutrophil Extracellular Traps in Fatal COVID-19-Associated Lung Injury. Dis. Markers 2021, 2021, 5566826. [Google Scholar] [CrossRef] [PubMed]
  144. Iwasawa, T.; Sato, M.; Yamaya, T.; Sato, Y.; Uchida, Y.; Kitamura, H.; Hagiwara, E.; Komatsu, S.; Utsunomiya, D.; Ogura, T. Ultra-high-resolution computed tomography can demonstrate alveolar collapse in novel coronavirus (COVID-19) pneumonia. Jpn. J. Radiol. 2020, 38, 394–398. [Google Scholar] [CrossRef]
  145. Lang, M.; Som, A.; Carey, D.; Reid, N.; Mendoza, D.P.; Flores, E.J.; Li, M.D.; Shepard, J.O.; Little, B.P. Pulmonary Vascular Manifestations of COVID-19 Pneumonia. Radiol. Cardiothorac. Imaging 2020, 2, e200277. [Google Scholar] [CrossRef]
  146. Grimmer, B.; Kuebler, W.M. The endothelium in hypoxic pulmonary vasoconstriction. J. Appl. Physiol. 2017, 123, 1635–1646. [Google Scholar] [CrossRef]
  147. D’Agnillo, F.; Walters, K.A.; Xiao, Y.; Sheng, Z.M.; Scherler, K.; Park, J.; Gygli, S.; Rosas, L.A.; Sadtler, K.; Kalish, H.; et al. Lung epithelial and endothelial damage, loss of tissue repair, inhibition of fibrinolysis, and cellular senescence in fatal COVID-19. Sci. Transl. Med. 2021, 13, eabj7790. [Google Scholar] [CrossRef]
  148. Batah, S.S.; Fabro, A.T. Pulmonary pathology of ARDS in COVID-19: A pathological review for clinicians. Respir. Med. 2021, 176, 106239. [Google Scholar] [CrossRef]
  149. Torres Acosta, M.A.; Singer, B.D. Pathogenesis of COVID-19-induced ARDS: Implications for an ageing population. Eur. Respir. J. 2020, 56, 2002049. [Google Scholar] [CrossRef]
  150. Helal, M.A.; Shouman, S.; Abdelwaly, A.; Elmehrath, A.O.; Essawy, M.; Sayed, S.M.; Saleh, A.H.; El-Badri, N. Molecular basis of the potential interaction of SARS-CoV-2 spike protein to CD147 in COVID-19 associated-lymphopenia. J. Biomol. Struct. Dyn. 2022, 40, 1109–1119. [Google Scholar] [CrossRef]
  151. Grifoni, A.; Weiskopf, D.; Ramirez, S.I.; Mateus, J.; Dan, J.M.; Moderbacher, C.R.; Rawlings, S.A.; Sutherland, A.; Premkumar, L.; Jadi, R.S.; et al. Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell 2020, 181, 1489–1501.e15. [Google Scholar] [CrossRef] [PubMed]
  152. Thompson, E.A.; Cascino, K.; Ordonez, A.A.; Zhou, W.; Vaghasia, A.; Hamacher-Brady, A.; Brady, N.R.; Sun, I.H.; Wang, R.; Rosenberg, A.Z.; et al. Metabolic programs define dysfunctional immune responses in severe COVID-19 patients. Cell Rep. 2021, 34, 108863. [Google Scholar] [CrossRef] [PubMed]
  153. Xiong, Y.; Liu, Y.; Cao, L.; Wang, D.; Guo, M.; Jiang, A.; Guo, D.; Hu, W.; Yang, J.; Tang, Z.; et al. Transcriptomic characteristics of bronchoalveolar lavage fluid and peripheral blood mononuclear cells in COVID-19 patients. Emerg. Microbes. Infect. 2020, 9, 761–770. [Google Scholar] [CrossRef] [PubMed]
  154. Chen, I.Y.; Moriyama, M.; Chang, M.F.; Ichinohe, T. Severe Acute Respiratory Syndrome Coronavirus Viroporin 3a Activates the NLRP3 Inflammasome. Front. Microbiol. 2019, 10, 50. [Google Scholar] [CrossRef]
  155. Paolini, A.; Borella, R.; De Biasi, S.; Neroni, A.; Mattioli, M.; Lo Tartaro, D.; Simonini, C.; Franceschini, L.; Cicco, G.; Piparo, A.M.; et al. Cell Death in Coronavirus Infections: Uncovering Its Role during COVID-19. Cells 2021, 10, 1585. [Google Scholar] [CrossRef]
  156. Gibellini, L.; De Biasi, S.; Paolini, A.; Borella, R.; Boraldi, F.; Mattioli, M.; Lo Tartaro, D.; Fidanza, L.; Caro-Maldonado, A.; Meschiari, M.; et al. Altered bioenergetics and mitochondrial dysfunction of monocytes in patients with COVID-19 pneumonia. EMBO Mol. Med. 2020, 12, e13001. [Google Scholar] [CrossRef]
  157. Silvin, A.; Chapuis, N.; Dunsmore, G.; Goubet, A.G.; Dubuisson, A.; Derosa, L.; Almire, C.; Henon, C.; Kosmider, O.; Droin, N.; et al. Elevated Calprotectin and Abnormal Myeloid Cell Subsets Discriminate Severe from Mild COVID-19. Cell 2020, 182, 1401–1418.e18. [Google Scholar] [CrossRef]
  158. Severe Covid, G.G.; Ellinghaus, D.; Degenhardt, F.; Bujanda, L.; Buti, M.; Albillos, A.; Invernizzi, P.; Fernandez, J.; Prati, D.; Baselli, G.; et al. Genomewide Association Study of Severe COVID-19 with Respiratory Failure. N. Engl. J. Med. 2020, 383, 1522–1534. [Google Scholar]
  159. Laing, A.G.; Lorenc, A.; Del Molino Del Barrio, I.; Das, A.; Fish, M.; Monin, L.; Munoz-Ruiz, M.; McKenzie, D.R.; Hayday, T.S.; Francos-Quijorna, I.; et al. A dynamic COVID-19 immune signature includes associations with poor prognosis. Nat. Med. 2020, 26, 1623–1635. [Google Scholar] [CrossRef]
  160. Diao, B.; Wang, C.; Tan, Y.; Chen, X.; Liu, Y.; Ning, L.; Chen, L.; Li, M.; Liu, Y.; Wang, G.; et al. Reduction and Functional Exhaustion of T Cells in Patients with Coronavirus Disease 2019 (COVID-19). Front. Immunol. 2020, 11, 827. [Google Scholar] [CrossRef]
  161. Gangaev, A.; Ketelaars, S.L.C.; Isaeva, O.I.; Patiwael, S.; Dopler, A.; Hoefakker, K.; De Biasi, S.; Gibellini, L.; Mussini, C.; Guaraldi, G.; et al. Identification and characterization of a SARS-CoV-2 specific CD8(+) T cell response with immunodominant features. Nat. Commun. 2021, 12, 2593. [Google Scholar] [CrossRef] [PubMed]
  162. Liu, C.; Martins, A.J.; Lau, W.W.; Rachmaninoff, N.; Chen, J.; Imberti, L.; Mostaghimi, D.; Fink, D.L.; Burbelo, P.D.; Dobbs, K.; et al. Time-resolved systems immunology reveals a late juncture linked to fatal COVID-19. Cell 2021, 184, 1836–1857.e22. [Google Scholar] [CrossRef] [PubMed]
  163. Peruzzi, B.; Bencini, S.; Capone, M.; Mazzoni, A.; Maggi, L.; Salvati, L.; Vanni, A.; Orazzini, C.; Nozzoli, C.; Morettini, A.; et al. Quantitative and qualitative alterations of circulating myeloid cells and plasmacytoid DC in SARS-CoV-2 infection. Immunology 2020, 161, 345–353. [Google Scholar] [CrossRef] [PubMed]
  164. Ehrchen, J.M.; Sunderkotter, C.; Foell, D.; Vogl, T.; Roth, J. The endogenous Toll-like receptor 4 agonist S100A8/S100A9 (calprotectin) as innate amplifier of infection, autoimmunity, and cancer. J. Leukoc. Biol. 2009, 86, 557–566. [Google Scholar] [CrossRef]
  165. Narasaraju, T.; Yang, E.; Samy, R.P.; Ng, H.H.; Poh, W.P.; Liew, A.A.; Phoon, M.C.; van Rooijen, N.; Chow, V.T. Excessive neutrophils and neutrophil extracellular traps contribute to acute lung injury of influenza pneumonitis. Am. J. Pathol. 2011, 179, 199–210. [Google Scholar] [CrossRef]
  166. Karki, R.; Sharma, B.R.; Tuladhar, S.; Williams, E.P.; Zalduondo, L.; Samir, P.; Zheng, M.; Sundaram, B.; Banoth, B.; Malireddi, R.K.S.; et al. Synergism of TNF-alpha and IFN-gamma Triggers Inflammatory Cell Death, Tissue Damage, and Mortality in SARS-CoV-2 Infection and Cytokine Shock Syndromes. Cell 2021, 184, 149–168.e17. [Google Scholar] [CrossRef]
  167. Li, S.; Zhang, Y.; Guan, Z.; Li, H.; Ye, M.; Chen, X.; Shen, J.; Zhou, Y.; Shi, Z.L.; Zhou, P.; et al. SARS-CoV-2 triggers inflammatory responses and cell death through caspase-8 activation. Signal Transduct. Target. Ther. 2020, 5, 235. [Google Scholar] [CrossRef]
  168. Nakamura, H.; Kinjo, T.; Arakaki, W.; Miyagi, K.; Tateyama, M.; Fujita, J. Serum levels of receptor-interacting protein kinase-3 in patients with COVID-19. Crit. Care 2020, 24, 484. [Google Scholar] [CrossRef]
  169. Ren, Y.; Shu, T.; Wu, D.; Mu, J.; Wang, C.; Huang, M.; Han, Y.; Zhang, X.Y.; Zhou, W.; Qiu, Y.; et al. The ORF3a protein of SARS-CoV-2 induces apoptosis in cells. Cell Mol. Immunol. 2020, 17, 881–883. [Google Scholar] [CrossRef]
  170. Polack, F.P.; Thomas, S.J.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Perez, J.L.; Perez Marc, G.; Moreira, E.D.; Zerbini, C.; et al. Safety and Efficacy of the BNT162b2 mRNA COVID-19 Vaccine. N. Engl. J. Med. 2020, 383, 2603–2615. [Google Scholar] [CrossRef]
  171. Baden, L.R.; El Sahly, H.M.; Essink, B.; Kotloff, K.; Frey, S.; Novak, R.; Diemert, D.; Spector, S.A.; Rouphael, N.; Creech, C.B.; et al. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N. Engl. J. Med. 2021, 384, 403–416. [Google Scholar] [CrossRef] [PubMed]
  172. Cohen, M.J.; Oster, Y.; Moses, A.E.; Spitzer, A.; Benenson, S.; Israeli-Hospitals 4th Vaccine Working Group. Association of Receiving a Fourth Dose of the BNT162b Vaccine with SARS-CoV-2 Infection among Health Care Workers in Israel. JAMA Netw Open 2022, 5, e2224657. [Google Scholar] [CrossRef] [PubMed]
  173. Barthwal, M.S.; Dole, S.; Sahasrabudhe, T. Management of COVID-19: A comprehensive and practical approach. Med. J. Armed Forces India 2022. [Google Scholar] [CrossRef]
  174. Hammond, J.; Leister-Tebbe, H.; Gardner, A.; Abreu, P.; Bao, W.; Wisemandle, W.; Baniecki, M.; Hendrick, V.M.; Damle, B.; Simon-Campos, A.; et al. Oral Nirmatrelvir for High-Risk, Nonhospitalized Adults with COVID-19. N. Engl. J. Med. 2022, 386, 1397–1408. [Google Scholar] [CrossRef] [PubMed]
  175. Jayk Bernal, A.; Gomes da Silva, M.M.; Musungaie, D.B.; Kovalchuk, E.; Gonzalez, A.; Delos Reyes, V.; Martin-Quiros, A.; Caraco, Y.; Williams-Diaz, A.; Brown, M.L.; et al. Molnupiravir for Oral Treatment of COVID-19 in Nonhospitalized Patients. N. Engl. J. Med. 2022, 386, 509–520. [Google Scholar] [CrossRef] [PubMed]
  176. Marconi, V.C.; Ramanan, A.V.; de Bono, S.; Kartman, C.E.; Krishnan, V.; Liao, R.; Piruzeli, M.L.B.; Goldman, J.D.; Alatorre-Alexander, J.; de Cassia Pellegrini, R.; et al. Efficacy and safety of baricitinib for the treatment of hospitalised adults with COVID-19 (COV-BARRIER): A randomised, double-blind, parallel-group, placebo-controlled phase 3 trial. Lancet Respir. Med. 2021, 9, 1407–1418. [Google Scholar] [CrossRef]
  177. Kalil, A.C.; Patterson, T.F.; Mehta, A.K.; Tomashek, K.M.; Wolfe, C.R.; Ghazaryan, V.; Marconi, V.C.; Ruiz-Palacios, G.M.; Hsieh, L.; Kline, S.; et al. Baricitinib plus Remdesivir for Hospitalized Adults with COVID-19. N. Engl. J. Med. 2021, 384, 795–807. [Google Scholar] [CrossRef]
  178. WHO Solidarity Trial Consortium; Pan, H.; Peto, R.; Henao-Restrepo, A.M.; Preziosi, M.P.; Sathiyamoorthy, V.; Abdool Karim, Q.; Alejandria, M.M.; Hernandez Garcia, C.; Kieny, M.P.; et al. Repurposed Antiviral Drugs for COVID-19-Interim WHO Solidarity Trial Results. N. Engl. J. Med. 2021, 384, 497–511. [Google Scholar] [CrossRef]
  179. Group, R.C.; Horby, P.; Lim, W.S.; Emberson, J.R.; Mafham, M.; Bell, J.L.; Linsell, L.; Staplin, N.; Brightling, C.; Ustianowski, A.; et al. Dexamethasone in Hospitalized Patients with COVID-19. N. Engl. J. Med. 2021, 384, 693–704. [Google Scholar]
  180. Investigators, R.-C.; Gordon, A.C.; Mouncey, P.R.; Al-Beidh, F.; Rowan, K.M.; Nichol, A.D.; Arabi, Y.M.; Annane, D.; Beane, A.; van Bentum-Puijk, W.; et al. Interleukin-6 Receptor Antagonists in Critically Ill Patients with COVID-19. N. Engl. J. Med. 2021, 384, 1491–1502. [Google Scholar] [CrossRef]
Figure 1. Findings on HRCT and histopathological specimens of the lungs in patients with AE-IPF. In a 78-year-old male patient with AE-IPF, HRCT at the initial diagnosis of IPF shows subpleural-predominant interstitial fibrosis, traction bronchiectasis (arrow) and honeycombing (arrowhead) ((A), from Oda et al. [1] with permission). HRCT at the onset of AE-IPF (12 months after the initial diagnosis) shows diffuse areas of GGA superimposed on underlying fibrotic opacities (B) [1]. Autopsy lung specimens were from another 52-year-old male patient with AE-IPF, who died on day 8 from the onset of AE- IPF. Hematoxylin and eosin staining shows diffuse alveolar damage with hyaline membrane ((C), from Tanaka et al. [2] with permission). Immunohistochemical staining of autopsy lung specimens shows that infiltrating alveolar neutrophils are positive for S100A8 (lower magnification in (D) and higher magnification in (E)), which is a calcium-binding protein produced and released mainly by activated neutrophils [2]. More neutrophils with S100A8 existed in the alveolar septa than in the alveolar space. Abbreviations: HRCT, high-resolution computed tomography; AE, acute exacerbation; IPF, idiopathic pulmonary fibrosis; GGA, ground-glass attenuation.
Figure 1. Findings on HRCT and histopathological specimens of the lungs in patients with AE-IPF. In a 78-year-old male patient with AE-IPF, HRCT at the initial diagnosis of IPF shows subpleural-predominant interstitial fibrosis, traction bronchiectasis (arrow) and honeycombing (arrowhead) ((A), from Oda et al. [1] with permission). HRCT at the onset of AE-IPF (12 months after the initial diagnosis) shows diffuse areas of GGA superimposed on underlying fibrotic opacities (B) [1]. Autopsy lung specimens were from another 52-year-old male patient with AE-IPF, who died on day 8 from the onset of AE- IPF. Hematoxylin and eosin staining shows diffuse alveolar damage with hyaline membrane ((C), from Tanaka et al. [2] with permission). Immunohistochemical staining of autopsy lung specimens shows that infiltrating alveolar neutrophils are positive for S100A8 (lower magnification in (D) and higher magnification in (E)), which is a calcium-binding protein produced and released mainly by activated neutrophils [2]. More neutrophils with S100A8 existed in the alveolar septa than in the alveolar space. Abbreviations: HRCT, high-resolution computed tomography; AE, acute exacerbation; IPF, idiopathic pulmonary fibrosis; GGA, ground-glass attenuation.
Ijms 23 15027 g001
Figure 2. HRCT findings of the lungs in a patient with DM-ILD with anti-MDA5 antibody. A forty-four-year-old male patient with DM-ILD was positive for anti-MDA5 antibody. At diagnosis, peripheral and peribronchovascular consolidations are observed (arrowheads). Interlobular septal thickening and non-septal linear or plate-like opacities are also seen (arrows) ((A,B), from Tanizawa et al. [92] with permission). Despite treatments for 6 weeks, severe respiratory failure developed, requiring mechanical ventilation. Diffuse GGA and consolidation with air bronchograms are extended in the whole lungs (C,D) [92]. Surveillance at this point revealed no evidence of infection. The patient died of respiratory failure one week later. Abbreviations: HRCT, high-resolution computed tomography; DM, dermatomyositis; ILD, interstitial lung disease; MDA, anti-melanoma differentiation-associated gene; GGA, ground-glass attenuation.
Figure 2. HRCT findings of the lungs in a patient with DM-ILD with anti-MDA5 antibody. A forty-four-year-old male patient with DM-ILD was positive for anti-MDA5 antibody. At diagnosis, peripheral and peribronchovascular consolidations are observed (arrowheads). Interlobular septal thickening and non-septal linear or plate-like opacities are also seen (arrows) ((A,B), from Tanizawa et al. [92] with permission). Despite treatments for 6 weeks, severe respiratory failure developed, requiring mechanical ventilation. Diffuse GGA and consolidation with air bronchograms are extended in the whole lungs (C,D) [92]. Surveillance at this point revealed no evidence of infection. The patient died of respiratory failure one week later. Abbreviations: HRCT, high-resolution computed tomography; DM, dermatomyositis; ILD, interstitial lung disease; MDA, anti-melanoma differentiation-associated gene; GGA, ground-glass attenuation.
Ijms 23 15027 g002
Figure 3. CT and histopathology of EGFR-TKI-induced lung injury. A 61-year-old male with lung adeno carcinoma received gefitinib. This case showed extensive diffuse GGA consistent with a DAD pattern on CT ((A), from Inoue et al. [4] with permission). Another 85-year-old male with lung squamous carcinoma and IPF received thoracic irradiation and gefitinib. A lung histopathological specimen with haematoxylin and eosin staining shows DAD pattern (B) [4]. This case died of rapidly progressive respiratory failure despite treatments with high-dose corticosteroids. Abbreviations: CT, computed tomography; EGFR-TKI, epidermal growth factor receptor-tyrosine kinase inhibitor; GGA, ground-glass attenuation; DAD, diffuse alveolar damage.
Figure 3. CT and histopathology of EGFR-TKI-induced lung injury. A 61-year-old male with lung adeno carcinoma received gefitinib. This case showed extensive diffuse GGA consistent with a DAD pattern on CT ((A), from Inoue et al. [4] with permission). Another 85-year-old male with lung squamous carcinoma and IPF received thoracic irradiation and gefitinib. A lung histopathological specimen with haematoxylin and eosin staining shows DAD pattern (B) [4]. This case died of rapidly progressive respiratory failure despite treatments with high-dose corticosteroids. Abbreviations: CT, computed tomography; EGFR-TKI, epidermal growth factor receptor-tyrosine kinase inhibitor; GGA, ground-glass attenuation; DAD, diffuse alveolar damage.
Ijms 23 15027 g003
Figure 4. HRCT of the lungs in COVID-19. On HRCT, multiple GGA, crazy paving patterns, and consolidation are seen in peripheral or peribronchiolar lung area in many cases with COVID-19. These findings on HRCT change over time by phases, such as multiple GGA in the early phase (A), crazy paving appearance 10 days after the onset of symptoms in the progressive to peak phase (B), and multifocal consolidation with mild parenchymal distortion another 7 days later in absorption phase (C) (from Larici et al. [142] with permission). CT showing the presence of “enlarging vessel” sign within the areas of increased lung density (arrows, (D)) [142]. Acute pulmonary embolism is seen in severe COVID-19 patients (arrowhead, (E,F)) [142]. Abbreviations: HRCT, high-resolution computed tomography; COVID-19, coronavirus disease 2019; GGA, ground-glass attenuation.
Figure 4. HRCT of the lungs in COVID-19. On HRCT, multiple GGA, crazy paving patterns, and consolidation are seen in peripheral or peribronchiolar lung area in many cases with COVID-19. These findings on HRCT change over time by phases, such as multiple GGA in the early phase (A), crazy paving appearance 10 days after the onset of symptoms in the progressive to peak phase (B), and multifocal consolidation with mild parenchymal distortion another 7 days later in absorption phase (C) (from Larici et al. [142] with permission). CT showing the presence of “enlarging vessel” sign within the areas of increased lung density (arrows, (D)) [142]. Acute pulmonary embolism is seen in severe COVID-19 patients (arrowhead, (E,F)) [142]. Abbreviations: HRCT, high-resolution computed tomography; COVID-19, coronavirus disease 2019; GGA, ground-glass attenuation.
Ijms 23 15027 g004
Table 1. Cells and molecules related with a pathogenesis of each disease.
Table 1. Cells and molecules related with a pathogenesis of each disease.
Alveolar Epithelial Cell InjuryLung Endothelial Cell InjuryActivated Alveolar MacrophageDCLymphocyteMonocyteActivated NeutrophilOther CellsHistopathological Patterns and Other Findings
AE-IPFKL-6↑, SP-D↑
α-defensin↑
Periostin↑
Corisin↑ (staphylococcus)
Pepsin↑, HMGB1↑
Apoptosis↑
HSP70↓
Thrombomodulin↓
CCL18↑
Ferritin↑
Hemosiderin score↑
HMGB1↑
DC↑Memory T cells↑ S100A8↑
S100A9↑
MMP9↑
IL-8↑
Adiponectin↑
Leptin↑
DAD
Pulmonary haemorrhage
Thromboembolism
CADM-ILDYKL-40↑ Anti-MDA5 ab↑
Ferritin↑, YKL-40↑
Chitotriosidase↑
CD163↑, CD206↑
CCL2↑
Interferon β↑
Hemophagocytic score↑
Lymphocyte↓
(H-Ferritin↑, TIM2)
Monocytes↓
(H-Ferritin↑, TIM2)
CCL2↑ IFIT3↑
Chitotriosidase↑
IL-8↑
DAD
EGFR-TKI-induced lung injury HSP70↓
Apoptosis↑
Mitochondrial ROS↑
NLRP3↑, IL-1β↑
HMGB1↑
Pyroptosis↑
Neutrophils↑ DAD
COVID-19 and ILDDirect invasion of virus
PANoptosis↑
Direct invasion of virus
PANoptosis↑
Thrombosis↑
Plasmacytoid DC↓
Myeloid DC↓
Direct invasion of virus
Lymphocytes↓
Mitochondrial ROS↑, NLRP3↑
PD-1↑, PDL-1↑
CXCR6↓, TNFα↑
IFNγ↑
PANoptosis↑
Monocytes↓
Mitochondrial damage↑
PD-1↑, PDL-1↑
S100A8↑
S100A9↑
NETs↑
DAD
Thrombosis
Abbreviations: AE, acute exacerbation; IPF, idiopathic pulmonary fibrosis; CADM, clinically amyopathic dermatomyositis; ILD, interstitial lung disease; EGFR-TKI, epidermal growth factor receptor-tyrosine kinase inhibitor; COVID-19, coronavirus disease 2019; DC, dendritic cell; KL-6, Krebs von den Lungen-6; SP-D, surfactant protein-D; HMGB1, high mobility group box protein1; HSP, heat shock protein; MMP, matrix metalloproteinase; DAD, diffuse alveolar damage; TIM, T-cell immunoglobulin and mucin domains; IFIT, interferon-induced protein with tetratricopeptide repeats; ROS, reactive oxygen species; NLRP, NLR family pyrin domain-containing protein; NETs, neutrophil extracellular traps. “↑” means the increased concentration or numbers of the indicated molecules or cells. “↓” means the decreased concentration or numbers of the indicated molecules or cells.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Enomoto, N. Pathological Roles of Pulmonary Cells in Acute Lung Injury: Lessons from Clinical Practice. Int. J. Mol. Sci. 2022, 23, 15027. https://doi.org/10.3390/ijms232315027

AMA Style

Enomoto N. Pathological Roles of Pulmonary Cells in Acute Lung Injury: Lessons from Clinical Practice. International Journal of Molecular Sciences. 2022; 23(23):15027. https://doi.org/10.3390/ijms232315027

Chicago/Turabian Style

Enomoto, Noriyuki. 2022. "Pathological Roles of Pulmonary Cells in Acute Lung Injury: Lessons from Clinical Practice" International Journal of Molecular Sciences 23, no. 23: 15027. https://doi.org/10.3390/ijms232315027

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