Skip to main content
Erschienen in: BMC Pulmonary Medicine 1/2019

Open Access 01.12.2019 | Research article

The overexpression of peroxiredoxin-4 affects the progression of idiopathic pulmonary fibrosis

verfasst von: Tetsuya Hanaka, Takashi Kido, Shingo Noguchi, Sohsuke Yamada, Hirotsugu Noguchi, Xin Guo, Aya Nawata, Ke-Yong Wang, Keishi Oda, Tsutomu Takaki, Hiroto Izumi, Hiroshi Ishimoto, Kazuhiro Yatera, Hiroshi Mukae

Erschienen in: BMC Pulmonary Medicine | Ausgabe 1/2019

Abstract

Background

Acute exacerbation of idiopathic pulmonary fibrosis (AE-IPF) is life-threatening. Several serum biomarkers, such as Krebs von den Lungen-6 (KL-6) and surfactant protein D (SP-D), are clinically used for evaluating AE-IPF, but these biomarkers are not adequate for establishing an early and accurate diagnosis of AE-IPF. Recently, the protective roles of the members of the peroxiredoxin (PRDX) family have been reported in IPF; however, the role of PRDX4 in AE-IPF is unclear.

Methods

Serum levels of PRDX4 protein, KL-6, SP-D and lactate dehydrogenase (LDH) in 51 patients with stable IPF (S-IPF), 38 patients with AE-IPF and 15 healthy volunteers were retrospectively assessed using enzyme-linked immunosorbent assay. Moreover, as an animal model of pulmonary fibrosis, wild-type (WT) and PRDX4-transgenic (Tg) mice were intratracheally administered with bleomycin (BLM, 2 mg/kg), and fibrotic and inflammatory changes in lungs were evaluated 3 weeks after the intratracheal administration.

Results

Serum levels of PRDX4 protein, KL-6, SP-D and LDH in patients with S-IPF and AE-IPF were significantly higher than those in healthy volunteers, and those in AE-IPF patients were the highest among the three groups. Using receiver operating characteristic curves, area under the curve values of serum PRDX4 protein, KL-6, SP-D, and LDH for detecting AE-IPF were 0.873, 0.698, 0.675, and 0.906, respectively. BLM-treated Tg mice demonstrated aggravated histopathological findings and poor prognosis compared with BLM-treated WT mice. Moreover, PRDX4 expression was observed in alveolar macrophages and lung epithelial cells of BLM-treated Tg mice.

Conclusions

PRDX4 is associated with the aggravation of inflammatory changes and fibrosis in the pathogenesis of IPF, and serum PRDX4 may be useful in clinical practice of IPF patients.
Hinweise

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12890-019-1032-2.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
8-OHdG
8-hydroxy-2’-deoxyguanosine
A-aDO2
Alveolar–arterial difference of oxygen
AE-IPF
Acute exacerbation of idiopathic pulmonary fibrosis
BALF
Bronchoalveolar lavage fluid
BLM
Bleomycin
CCL-2
CC chemokine ligand 2
CTGF
Connective tissue growth factor
DLCO
Diffusing capacity of the lungs for carbon monoxide
FVC
Forced vital capacity
GAPDH
Glyceraldehyde 3-phosphate dehydrogenase
HE
Hematoxylin and eosin
IFN-γ
Interferon γ
IL
Interleukin
IPF
Idiopathic pulmonary fibrosis
KL-6
Krebs von den Lungen-6
PaO2/FiO2
Arterial partial pressure of oxygen/fraction of inspired oxygen
PCR
Polymerase chain reaction
PDGF-B
Platelet-derived growth factor subunit B
PDGFRs
Platelet-derived growth factor receptors
PF
Pulmonary fibrosis
PRDX
Peroxiredoxin
PRDX4
Peroxiredoxin 4
S-IPF
Stable idiopathic pulmonary fibrosis
SP-D
Surfactant protein D
TGF-β1
Active tissue growth factor-β1
TNF-α
Tumor necrosis factor-α
TTF-1
Thyroid transcription factor
UIP
Usual interstitial pneumonia
α-SMA
α-smooth muscle actin

Background

Idiopathic pulmonary fibrosis (IPF) is a fibrosing parenchymal lung disease that has chronic, progressive, and even fatal outcomes. The natural history of patients with IPF is extremely complex; moreover, the clinical course of such patients varies from relatively stable to sudden acute exacerbation, which often prove fatal. Recently, clinical trials have demonstrated that antifibrotic agents, such as pirfenidone and nintedanib, reduce the decline in forced vital capacity (FVC) and prolong progression-free survival in patients with IPF [1, 2]; however, the prognosis of IPF is still poor with an estimated median survival time of 2–5 years after the initial diagnosis [3].
Approximately 5–10% of patients with IPF generally demonstrate acute exacerbation (AE) annually [4, 5], leading to very high mortality [6]. An early diagnosis of AE-IPF is the most important clinical parameter; however, available data regarding useful biomarkers to precisely predict progressive patients with IPF are limited. Therefore, simple and effective diagnostic biomarkers for detecting AE-IPF are required for prompt decision making in the proper treatment of patients with IPF. Several serum biomarkers, such as Krebs von den Lungen-6 (KL-6) and surfactant proteins A and D (SP-A and -D, respectively), are clinically used for diagnosing AE-IPF [7, 8], but these markers are not adequately efficient; other biomarkers, such as monomeric periostin [9] and heat shock protein 47 (HSP47) [10], have also been recently reported as surrogate markers for detecting AE-IPF.
Peroxiredoxin (PRDX) is a recently identified antioxidant family that contains reactive cysteine in a conserved region near the N-terminus [11, 12]. Six members of the PRDX family have been identified in mammals (PRDX1–6). Human PRDX4 is the only secretory isoform that exists in both intra- and extracellular spaces [13, 14] and is ubiquitously synthesized and is abundantly expressed in various organisms [15]. Regarding the role of each PRDX in IPF, a protective role of PRDX1 in bleomycin (BLM)-induced pulmonary fibrosis (PF) mice was reported [16]. In addition, the co-localization of PRDX2 with platelet-derived growth factor receptors (PDGFRs) and proliferating cells in human lung tissue in patients with IPF/usual interstitial pneumonia (UIP) was also reported [17]. In addition, an increase in the PRDX4 mRNA expression in the lung tissue of patients with interstitial lung disease was reported [18], but the role of PRDX4 in the pathogenesis and progression of IPF is still unclear.
We previously generated human PRDX4-transgenic mice (Tg mice) using C57BL/6 mice and reported that PRDX4 may have a protective role against the progression of atherosclerosis and nonalcoholic fatty liver disease via its antioxidant effect [19, 20].
In the present study, we compared the serum PRDX4 protein level in patients with stable IPF (S-IPF), AE-IPF and healthy volunteers to evaluate the significance of PRDX4 in patients with IPF. In addition, we examined the pathogenetic roles of PRDX4 in pulmonary inflammation and fibrosis using Tg mice in a BLM-induced PF model.

Methods

Human study

S-IPF and AE-IPF were diagnosed based on the criteria for IPF and AE-IPF [4, 21, 22], respectively. The serum samples obtained from patients with S-IPF and AE-IPF between April 2010 and December 2016 were analyzed for serum PRDX4 protein level. In addition, serum samples of 15 healthy adult volunteers (32–47 years old) with no medical histories were also collected. This study was conducted according to the Declaration of Helsinki and was approved by the Ethics Committee of Medical Research, University of Occupational and Environmental Health, Japan (approval number H29–182). For patients with S-IPF in whom AE-IPF occurred during the follow-up period, the serum samples were obtained both in the stable state of IPF (as S-IPF) prior to AE-IPF and at the time of AE of IPF (as AE-IPF). Clinical data such as age, sex, body mass index (BMI), and smoking history; clinical manifestations; and laboratory data including serum KL-6, SP-D, and lactate dehydrogenase (LDH) levels were collected.

Serum and bronchoalveolar lavage fluid (BALF) PRDX4 protein levels in humans and mice

Serum and BALF PRDX4 protein levels in both humans and mice were assessed using enzyme-linked immunosorbent assay (ELISA) (Abnova, Taipei, Taiwan) according to the manufacturer’s protocol as previously described [20].

Animal study

The animal study was approved by the Ethics Committee of Animal Care and Experimentation, University of Occupational and Environmental Health, Japan (approval number AE-14-019) and performed in accordance with the National Institutes of Health guidelines. Male wild-type (WT) mice (C57BL/6, 10-week-old) and Tg mice (weight, 21–28 g) were selected and maintained on a regular diet (CE-2, CLEA Japan, Inc., Tokyo, Japan). WT mice were obtained from Kyudo Co., Ltd. (Tosu, Japan). PRDX4-Tg mice were generated and provided in our facility [23].

Intratracheal BLM treatment in mice

2.0-mg/kg BLM (Nippon Kayaku, Tokyo, Japan) was dissolved in 40-μL sterile saline. This solution (BLM group) or 40-μL sterile saline alone (saline group) was intratracheally instilled in both WT and Tg mice after receiving intraperitoneal sodium pentobarbital. Body weights were recorded at 0, 3, 7, 14 and 21 days after the intratracheal instillation. On day 21, the mice were inhaled with 3% sevoflurane to initiate anesthesia and then deeply anesthetized with intraperitoneal injection of sodium pentobarbital (50 mg/kg). These mice were euthanized by cutting the inferior vena cava to induce exsanguination after collecting blood from the inferior vena cava. Separately, a group sacrificed on day 0 without any intratracheal instillation was provided in order to evaluate the human PRDX4 mRNA and protein levels at baseline (baseline group).
The numbers of mice were as follows: WT mice at baseline (WT-baseline) (n = 5), Tg mice at baseline (Tg-baseline) (n = 5), saline-treated WT mice (WT-saline) (n = 5), saline-treated Tg mice (Tg-saline) (n = 5), BLM-treated WT mice (WT-BLM) (n = 14) and BLM-treated Tg mice (Tg-BLM) (n = 14). The numbers of mice that survived until day 21 among the saline- and BLM-treated mice were as follows: WT-saline (n = 5), Tg-saline (n = 5), WT-BLM (n = 12) and Tg-BLM (n = 7). Furthermore, to avoid the influence of BAL on other experimental results, the following numbers of mice were prepared for the BALF analysis: WT-baseline (n = 5), Tg-baseline (n = 5), WT-saline (n = 5), Tg-saline (n = 5), WT-BLM (n = 14) and Tg-BLM (n = 14). The number of mice that survived until day 21 among the saline- and BLM-treated mice for the BALF analysis were as follows: WT-saline (n = 5), Tg-saline (n = 5), WT-BLM (n = 12) and Tg-BLM (n = 6).

Microscopic computed tomography in mice

Under general anesthesia induced by inhaling sevoflurane, microscopic computed tomography (micro-CT) images of mouse lungs were evaluated on day 21 after instillation using a micro-CT system (CosmoScan GX, Rigaku Co., Tokyo, Japan) using the following conditions: 90 kV, 88 μA; field of view, 36 mm; voxel size, 60 × 60 × 60 μm; and scan time, 4 min.

BALF in mice

BALF in mice was obtained by cannulating the trachea using a 20-gage catheter and by washing three times using 1-mL sterile saline. Cytospin was performed to evaluate the presence of BALF cells, and the obtained cell-free supernatants were stored at − 80 °C until PRDX4 protein assessment as previously described [20].

Histopathological and immunostaining assessments of murine lungs

Left lungs of the mice were removed by incising at the anterior midline, were fixed with 15% formalin neutral buffer solution (Wako, Osaka, Japan) at 25 cmH2O, and were embedded in paraffin. Subsequently, 3-μm sections of embedded lung tissues were stained with hematoxylin and eosin (HE) and Masson’s trichrome. The Ashcroft score was assessed to evaluate PF as previously described [24]. Each specimen was independently scored by two observers (TH and WKY), including a histopathologist, and the mean scores were considered as the fibrotic score.
The mouse anti-human monoclonal fibronectin antibody (1:100; Abcam, Cambridge, United Kingdom), rabbit anti-human PRDX4 polyclonal antibody (1:500; BioReagents, Golden, CO, USA) [19, 23, 25], and anti-mouse monoclonal antibody for 8-hydroxy-2′-deoxyguanosine (8-OHdG) (1:100; Japan Institute for the Control of Aging, Fukuroi, Japan) were used. The number of positively stained cells in five randomly selected fields per section was quantified (original magnification: × 200) as previously described [19, 23] in analyses of 8-OHdG.

Double immunofluorescence staining of murine lungs

For immunofluorescence studies, the sections of right lung of Tg mice were embedded in the OCT compound (Sakura Finetek Japan, Tokyo, Japan), snap-frozen in liquid nitrogen, and stored at − 80 °C until use. To identify PRDX4-positive cells in lung tissues, 6-μm-thick cryosections were used for double immunofluorescence staining of polyclonal rabbit anti-human PRDX4 antibody (1:500; Thermo Fisher Scientific, Yokohama, Japan) and were visualized using goat anti-rabbit IgG antibodies conjugated with Alexa Fluor 488® (green; Thermo Fisher Scientific, Yokohama, Japan) combined with monoclonal rat anti-mouse Mac-2 (1:500; Cedarlane Laboratories, Burlington, Canada), monoclonal mouse anti-rat thyroid transcription factor (TTF-1; 1:100; Dako Cytomation Co., Tokyo, Japan), and monoclonal mouse anti-human α-smooth muscle actin (α-SMA; 1:150; Dako Cytomation Co., Tokyo, Japan) antibodies visualized using goat anti-mouse IgG antibodies conjugated with Alexa Fluor 546® (red; Thermo Fisher Scientific, Yokohama, Japan).

Real-time polymerase chain reaction (PCR)

The total RNA extracted from the homogenized right lung tissue using ISOGEN reagent (Nippon Gene, Tokyo, Japan) was reverse-transcribed. The expression of CC chemokine ligand 2 (CCL-2), collagen 1A1, connective tissue growth factor (CTGF), human PRDX4, interferon γ (IFN-γ), interleukin (IL)-1β, IL-4, IL-6, IL-13, IL-17A, tumor necrosis factor (TNF)-α, platelet-derived growth factor subunit B (PDGF-B), active tissue growth factor-β1 (TGF-β1), fibronectin and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was quantified using real-time quantitative PCR with the ABI prism 7000 sequence detection system (Applied Biosystems, Foster City, CA, USA), as previously described [20, 26]. The relative expression level of each gene was normalized to that of GAPDH using random primers as previously reported [27].

Fibronectin protein concentration in murine lungs

Fibronectin protein concentrations in mouse lung homogenates were measured using the ELISA kit (Abbexa, Cambridge, United Kingdom) according to the manufacturer’s protocol.

Statistical analysis

Data are presented as medians (interquartile range) or the number of subjects (%) in human data and as means (standard error of the mean) in murine data, unless otherwise specified. Continuous variables were compared using the Mann–Whitney U test or Kruskal-Wallis test, and categorical variables were compared using the chi-square test or Fisher’s exact test, as appropriate. Changes in human serum PRDX4 protein, KL-6, SP-D, and LDH levels were determined using the Wilcoxon signed rank test. The receiver operating characteristic (ROC) curves and Youden indices were used to determine the optimal cut-off of serum PRDX4, KL-6, SP-D, and LDH levels to distinguish AE-IPF from S-IPF. The survival probability of each group was estimated using the Kaplan–Meier method and compared using the global log-rank test. Values of P <  0.05 were considered to be statistically significant. All calculations were performed using the StatFlex software version 6 (Artech, Osaka, Japan).

Results

Clinical characteristics of patients with IPF

We analyzed the blood samples of 51 patients with S-IPF, 38 patients with AE-IPF and 15 healthy volunteers. Among them, BALF samples were also evaluated in 14 S-IPF and 10 AE-IPF patients. During the observation period, 9 of the 51 S-IPF patients developed AE, and the serum samples of these 9 patients were collected in both the stable state before AE developed (as S-IPF) and also at time of AE development (as AE-IPF). Table 1 shows the characteristics of the enrolled patients. Compared with the S-IPF group, the AE-IPF group showed a significantly lower arterial partial pressure of oxygen/fraction of inspired oxygen (PaO2/FiO2) ratio, significantly higher alveolar–arterial difference of oxygen (A-aDO2), significantly lower FVC and significantly lower diffusing capacity of the lungs for carbon monoxide (DLCO), and the overall survival of the AE-IPF group was significantly worse than that of the S-IPF group after the diagnosis.
Table 1
Characteristics of healthy volunteers and patients with S-IPF and AE-IPF
 
HV (n = 15)
S-IPF (n = 51)
AE-IPF (n = 38)
Three groups, p-value
HV/S-IPF, p-value
HV/AE-IPF, p-value
S-IPF/AE-IPF, p-value
Age, years
36 (34–40)
72 (68–75)
72 (64–75)
<  0.001
<  0.001
<  0.001
0.983
Sex, male
13 (87)
39 (76)
35 (92)
0.217
0.396
0.542
0.051
BMI
22.9 (20.9–24.8)
22.9 (20.7–24.7)
22.9 (21.1–25.8)
0.785
0.786
0.875
0.480
Smoking history, never/former/current
15/0/0
12/30/9
2/31/5
<  0.001
<  0.001
<  0.001
0.254
PaO2/FiO2 ratio, mmHg
395 (324–429)
220 (128–296)
<  0.001
A-a DO2, mmHg
14 (9–31)
135 (58–251)
<  0.001
FVCa, L
2.00 (1.43–2.66)
1.85 (1.73–2.31)
0.674
FVCa, % predicted
67 (55–79)
54 (47–68)
0.039
DLCOb, % predicted
66 (43–83)
37 (29–49)
0.005
Overall survivalc, days
1059
57
<  0.001
Data are presented as median (interquartile range) or n (%)
aThe numbers of patients with FVC measured were as follows: S-IPF (n = 51), AE-IPF (n = 15)
bThe numbers of patients with DLCO measured were as follows: S-IPF (n = 35), AE-IPF (n = 10)
cThe date of baseline of overall survival is the day of diagnosis of S-IPF and AE-IPF
Abbreviations: AE-IPF Acute exacerbation of idiopathic pulmonary fibrosis, A-a DO2 Alveolar-arterial difference of oxygen, BMI Body mass index, DLCO Diffusing capacity of the lungs for carbon monoxide, FVC Forced vital capacity, HV Healthy volunteer, PaO2/FiO2 Arterial partial pressure of oxygen/fraction of inspired oxygen, S-IPF Stable idiopathic pulmonary fibrosis

Comparison of serum PRDX4 protein, KL-6, SP-D, and LDH levels

The serum levels of PRDX4 protein, KL-6, SP-D and LDH in patients with S-IPF were all significantly higher than those in healthy volunteers (Fig. 1), and those in AE-IPF patients were all significantly higher than those in S-IPF patients. In contrast, the BALF PRDX4 protein level did not differ significantly between the patients with S-IPF and AE-IPF (Additional file 1: Figure S1), nor were any significant correlations noted between the serum and BALF PRDX4 protein levels in either case (Additional file 2: Figure S2).

Changes in serum PRDX4 protein, KL-6, SP-D, and LDH levels in patients with S-IPF that subsequently progressed to AE-IPF

Nine patients with S-IPF subsequently progressed to AE-IPF, and the interval until the diagnosis from S-IPF to AE-IPF ranged from 62 to 1373 (median: 552) days. For these patients, changes in serum PRDX4 protein, KL-6, SP-D, and LDH levels at S-IPF and AE-IPF were compared. Serum PRDX4 protein levels at AE-IPF were significantly higher than those at S-IPF (p <  0.05) (Fig. 2a); however, serum KL-6, SP-D, and LDH levels showed no significant changes (Figs. 2b, c, and d).

The significance of serum PRDX4 protein, KL-6, SP-D, and LDH levels in AE-IPF using ROC curves

The area under the curves (AUCs) of serum PRDX4 protein, KL-6, SP-D, and LDH in AE-IPF were 0.873, 0.698, 0.675, and 0.906, respectively (Fig. 3). The optimal cut-off levels (Youden index) of serum PRDX4 protein, KL-6, SP-D, and LDH were 5.84 ng/mL, 1046 U/mL, 374 ng/mL, and 281 U/L, respectively. The sensitivities and specificities of serum PRDX4 protein, KL-6, SP-D, and LDH levels were 0.763 0.676, 0.556, and 0.763 and 0.961, 0.647, 0.740, and 0.961, respectively.

Survival rates and body weight changes after BLM treatment in mice

Figure 4a shows the survival time of mice until day 21 after the intratracheal administration of BLM or saline. The survival time of Tg-BLM was significantly lower than that of WT-BLM (p = 0.042). In addition, the body weights of Tg-BLM significantly decreased compared with those of WT-BLM (Fig. 4b).

Human PRDX4 expression in the murine lung

The immunohistochemistry of PRDX4 demonstrated the presence of PRDX4-positive cells in WT and Tg mice. The intensity and distribution of PRDX4 in Tg-BLM were greater than those in WT-BLM (Fig. 5a).
Double immunofluorescence staining of the lung sections of Tg mice (Fig. 5b) demonstrated that human PRDX4 and Mac-2, human PRDX4 and TTF-1 were expressed in the same cells in Tg-saline and Tg-BLM. Moreover, human PRDX4 and α-SMA were expressed in the same cells in Tg-BLM. These results suggest that human PRDX4 is mainly localized in alveolar macrophages and epithelial cells in the lungs of Tg mice.
In addition, the BALF human PRDX4 protein level in Tg-BLM was significantly higher than in WT-BLM (Fig. 5c). The serum human PRDX4 protein level in Tg-baseline, Tg-saline and Tg-BLM was significantly higher than in the WT mice (Fig. 5d). Furthermore, the human PRDX4 mRNA levels in lungs of Tg-baseline, Tg-saline and Tg-BLM were significantly higher than in the WT mice (Fig. 5e).

Quantification of PF in murine lungs

Representative pathological findings from murine lung tissues following HE, Masson’s trichrome, and immunohistochemical staining with anti-fibronectin antibody 21 days after BLM or saline treatment are shown in Fig. 6a. HE staining of lung tissues of both WT-saline and Tg-saline was normal whereas that of lung tissues of WT-BLM and Tg-BLM demonstrated pulmonary fibrotic changes in both WT and Tg mice. Masson’s trichrome and immunohistochemical staining revealed more intense collagen deposition and strong immunostaining of fibronectin in Tg-BLM than in WT-BLM (Fig. 6a). Chest micro-CT demonstrated traction bronchiectasis and severe consolidations in Tg-BLM compared with those in WT-BLM (Fig. 6b).
Ashcroft scores in both WT-saline (0.2 ± 0.0) and Tg-saline (0.2 ± 0.1) were almost normal; however, these scores in Tg-BLM were significantly higher than those in WT-BLM (4.9 ± 0.4 vs. 3.1 ± 0.6, respectively; p = 0.044) (Fig. 6c).
Both protein and mRNA levels of fibronectin in lung homogenates of Tg-BLM were significantly higher than those in lung homogenates of WT-BLM (Figs. 6d and e).

Quantitative analysis of proinflammatory cytokines and growth or profibrotic factor expressions in murine lungs

The IL-13 mRNA expression in the lung was significantly higher in Tg-saline than in WT-saline mice, but no marked differences were noted in the IL-1β, IL-6, TNF-α, CCL-2, IL-17A, TGF- β1, CTGF, PDGF-B or collagen 1A1 expression. In Tg-BLM mice, the pulmonary mRNA levels of CCL-2 and IL-17 were significantly higher while those of IL-4 and IFN-γ were significantly lower than in WT-BLM mice. However, no significant differences were found in other assessed cytokines or in growth or profibrotic factors between these two groups (Fig. 7).

Oxidative stress analysis

Immunohistochemical staining of murine lung tissues (Fig. 8a) demonstrated that the proportion of 8-OHdG-positive cells significantly increased in the BLM-treated group compared with that in the saline-treated group of both WT and Tg mice. However, the proportion of 8-OHdG-positive cells in WT-BLM and Tg-BLM was not significantly different (Fig. 8b).

Discussion

In the present study, we investigated the role and significance of PRDX4 in PF. In human study, the patients with AE-IPF had higher serum PRDX4 protein levels than those with S-IPF, and serum PRDX4 protein levels showed higher prognostic value than both serum KL-6 and SP-D levels for distinguishing AE-IPF from S-IPF. In addition, animal studies using a BLM-induced PF model of Tg mice demonstrated that Tg-BLM had significantly worse pulmonary fibrotic changes with significantly poor survival rates compared with WT-BLM mice.
Generally, patients with IPF are well known to exhibit elevated serum KL-6, SP-D, and LDH levels, and these levels are also recognized as predictive biomarkers of survival of these patients [4]. However, a Japanese report showed that the AUCs for serum KL-6, SP-D, and LDH levels for distinguishing AE-IPF from S-IPF were 0.576, 0.718, and 0.84, respectively [28]. Moreover, other reports showed that serum KL-6 and SP-D levels did not differ between patients with S-IPF and AE-IPF [10, 29], suggesting poor presence of potential biomarkers for distinguishing AE-IPF from S-IPF [10]. Similar to previous reports [10, 29], serum KL-6, SP-D, and LDH levels at AE-IPF were not significantly higher than those at S-IPF in this study. Conversely, serum PRDX4 protein and LDH levels showed better AUC profiles, obtained using ROC curves, to distinguish AE-IPF from S-IPF than serum KL-6 and SP-D levels. A recent report demonstrated that HSP47 (47 kDa) may be a potential biomarker for distinguishing AE-IPF from S-IPF, and monomeric periostin (90 kDa) and latent TGF-β binding protein-2 (195–240 kDa) are useful for predicting poor prognosis in patients with IPF [9, 30]. The molecular weight of these biomarkers is smaller than that of KL-6 (> 200 kDa) [31], and considering that the molecular weight is associated with biomarker profiles [10, 29], the small molecular weight of PRDX4 (34 kDa) [20] may explain its better profile as a marker for detecting AE-IPF. However, the molecular size of SP-D is 43 kDa, which is not considerably different from that of PRDX4. Recently, monomeric periostin was reported to be expressed in fibroblastic foci, and KL-6 and SP-D were expressed in regenerating alveolar type II cells, and these different release sites may partly explain the profiles of these biomarkers [9]. Further investigations are therefore warranted to elucidate the release sites of PRDX4 in patients with IPF.
PRDX4 is widely expressed in various organs other than the lungs. Moreover, elevated serum PRDX4 protein levels are associated with poor outcomes and high mortalities in patients with sepsis [32, 33]. The role of PRDX4 in pulmonary inflammation and fibrosis is still unclear, and the specificity of serum PRDX4 in patients with AE-IPF as a biomarker could not be evaluated in this study. However, increased serum PRDX4 level was associated with an aggravation of pulmonary inflammatory changes, fibrosis, and poor prognosis in the murine model; therefore, elevated serum PRDX4 levels observed in patients with AE-IPF may originate from increased PRDX4 expression in the lungs. Further studies are necessary to elucidate the clinical significance of serum PRDX4 levels in AE-IPF and other respiratory disorders.
In the present study, the mRNA levels of proinflammatory cytokines such as IL-1β, IL-6 and TNF-α in murine lung homogenates showed no significant differences between WT- and Tg-BLM. These proinflammatory cytokines are generally used as markers of the acute phase of PF in BLM-induced PF murine model [34], and these results may be influenced by the timing of evaluation. In contrast, significantly increased gene expressions of IL-17A and fibronectin were observed in the lung tissues of Tg-BLM compared with those in the lung tissues of WT-BLM. Reportedly, IL-17A is involved in the pathogenesis of BLM-induced PF [35], and early IL-17A axis leads to pulmonary inflammation and fibrosis in the late phase [36]. In addition, danger signals (damage-associated molecular patterns), which induce an immune response by acting on the dendritic cells, also cause tissue injury and inflammation that are mediated by IL-17A [37, 38]. Therefore, the PRDX4-induced overexpression of IL-17A may play an important role in the pathogenesis and progression of BLM-induced pulmonary inflammation and fibrosis. The expression of NF-κB-regulated cytokines in the WT and Tg mice after saline or BLM challenge was not markedly different in the present study, but both the suppressive effect of PRDX4 on NF-κB [13] and the activating effect of IL-17A on NF-κB [39] might partly explain the conflicting findings seen in the expression of NF-κB-regulated cytokines in the WT and Tg mice after saline or BLM treatment.
The oxidant–antioxidant imbalance plays an important role in the pathogenesis of IPF [40], and an increased expression of 8-OHdG, a marker of oxidative stress, is observed in the lungs of patients with IPF [41]. The expressions of 8-OHdG in the lungs of Tg-BLM and WT-BLM were not different in the present study. Kikuchi et al. reported that mice lacking PRDX1, a member of the PRDX family, exhibited aggravated lung inflammation and fibrosis due to an increase in the pulmonary oxidant stress [16]; moreover, Wang et al. demonstrated that a lack of PRDX6 resulted in lung injury in mice [42]. In addition, we previously reported regarding the antioxidant effects of PRDX4 in Tg mice in the models of diabetes mellitus [23], atherosclerosis [16], and nonalcoholic fatty liver disease [25]. Conversely, extracellular PRDXs, such as PRDX1, PRDX2, PRDX5 and PRDX6 induce severe inflammation in the brain by functioning as danger signals in brain injury models [38]; thus, conflicting actions of PRDXs have been reported in several inflammatory diseases. Although the mechanisms of the protective roles of PRDX4 are still unknown, stimulated danger signals including inflammatory cytokines, such as IL-6 and IL-8, other than profibrotic cytokines may play an important role in the pathogenesis and progression of AE in patients with IPF [43]. Our results suggest that the overexpression of PRDX4 in the lung may exert an exacerbating effect on pulmonary fibrosis by inducing inflammatory cytokines as danger signals rather than a protective effect as an antioxidant enzyme in the acute to subacute phase of pulmonary inflammation; however, further investigation regarding this is necessary.
Among the members of the PRDX family, PRDX1 is expressed in alveolar macrophages in the BLM-induced PF murine model [16]. However, the PRDX4 expression in the normal and inflamed human lungs is still unclear, and the types of cells that secrete PRDX4 as well as the ratio of secretion and intracellular PRDX4 in each cell type have been unclear in patients with IPF. In the present study, immunohistochemistry of murine lungs demonstrated the PRDX4 expression in alveolar macrophages and alveolar epithelial cells in Tg-BLM, although the amount and ratio of secreted and intracellular PRDX4 in each cell type remained unclear. This location is similar to that of the PRDX1 expressed in fibrotic murine lungs [16].
This study has several limitations. First, the human study was a single-center retrospective study with a limited number of patients with S-IPF and AE-IPF for detecting serum and BALF PRDX4 protein levels. Second, the backgrounds such as age, gender and smoking histories of the healthy volunteers and those of IPF patients were not matched. Third, we were unable to assess changes between baseline and follow-up period in pulmonary function, because many patients did not undergo a pulmonary function test during the follow-up period, therefore, we could not evaluate the relationship between serum PRDX4 and the change in pulmonary function. Fourth, cross-reaction of the anti-human PRDX4 antibody with mouse PRDX4 can be observed as the amino-acid sequences of human and mouse PRDX4 are highly homologous [25]; therefore, immunohistochemical staining of lungs of WT mice revealed human PRDX4-positive cells. Eventually, only male mice were used in this study, similar to our previous research [19, 20, 23, 25], and we were unable to evaluate the gender differences in the pathogenesis of IPF in Tg mice.

Conclusions

The results of the present study suggest that PRDX4 is associated with the aggravation of IPF and serum PRDX4 may be useful in clinical practice of IPF patients. Further studies are warranted to enable a better understanding of the detailed role of PRDX4 in IPF.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12890-019-1032-2.

Acknowledgements

Not applicable.
This study on humans was conducted according to guidelines of the Declaration of Helsinki and approved by the Ethics Committee of the Medical Research, University of Occupational and Environmental Health, Japan (approval number H29–182, October 10, 2017). Informed consent was waived because of the retrospective nature of the study. This study on animals was approved by the Ethics Committee of Animal Care and Experimentation, University of Occupational and Environmental Health, Japan (approval number AE-14-019, December 15, 2014) and was performed in accordance with the National Institutes of Health guidelines.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Schaffer JM, Singh SK, Reitz BA, Zamanian RT, Mallidi HR. Single- vs double-lung transplantation in patients with chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis since the implementation of lung allocation based on medical need. JAMA. 2015;313:936–48.PubMedCrossRef Schaffer JM, Singh SK, Reitz BA, Zamanian RT, Mallidi HR. Single- vs double-lung transplantation in patients with chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis since the implementation of lung allocation based on medical need. JAMA. 2015;313:936–48.PubMedCrossRef
2.
Zurück zum Zitat Rosas IO, Goldberg HJ, Collard HR, El-Chemaly S, Flaherty K, Hunninghake GM, et al. A phase II clinical trial of low-dose inhaled carbon monoxide in idiopathic pulmonary fibrosis. Chest. 2018;153:94–104.PubMedCrossRef Rosas IO, Goldberg HJ, Collard HR, El-Chemaly S, Flaherty K, Hunninghake GM, et al. A phase II clinical trial of low-dose inhaled carbon monoxide in idiopathic pulmonary fibrosis. Chest. 2018;153:94–104.PubMedCrossRef
3.
Zurück zum Zitat du Bois RM. An earlier and more confident diagnosis of idiopathic pulmonary fibrosis. Eur Respir Rev. 2012;21:141–6.PubMedCrossRef du Bois RM. An earlier and more confident diagnosis of idiopathic pulmonary fibrosis. Eur Respir Rev. 2012;21:141–6.PubMedCrossRef
4.
Zurück zum Zitat Raghu G, Collard HR, Egan JJ, Martinez FJ, Behr J, Brown KK, 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.PubMedPubMedCentralCrossRef Raghu G, Collard HR, Egan JJ, Martinez FJ, Behr J, Brown KK, 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.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Oda K, Yatera K, Fujino Y, Ishimoto H, Nakao H, Hanaka T, et al. Efficacy of concurrent treatments in idiopathic pulmonary fibrosis patients with a rapid progression of respiratory failure: an analysis of a national administrative database in Japan. BMC Pulm Med. 2016;16:91.PubMedPubMedCentralCrossRef Oda K, Yatera K, Fujino Y, Ishimoto H, Nakao H, Hanaka T, et al. Efficacy of concurrent treatments in idiopathic pulmonary fibrosis patients with a rapid progression of respiratory failure: an analysis of a national administrative database in Japan. BMC Pulm Med. 2016;16:91.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Juarez MM, Chan AL, Norris AG, Morrissey BM, Albertson TE. Acute exacerbation of idiopathic pulmonary fibrosis-a review of current and novel pharmacotherapies. J Thorac Dis. 2015;7:499–519.PubMedPubMedCentral Juarez MM, Chan AL, Norris AG, Morrissey BM, Albertson TE. Acute exacerbation of idiopathic pulmonary fibrosis-a review of current and novel pharmacotherapies. J Thorac Dis. 2015;7:499–519.PubMedPubMedCentral
7.
Zurück zum Zitat Takahashi H, Fujishima T, Koba H, Murakami S, Kurokawa K, Shibuya Y, et al. Serum surfactant proteins a and D as prognostic factors in idiopathic pulmonary fibrosis and their relationship to disease extent. Am J Respir Crit Care Med. 2000;162:1109–14.PubMedCrossRef Takahashi H, Fujishima T, Koba H, Murakami S, Kurokawa K, Shibuya Y, et al. Serum surfactant proteins a and D as prognostic factors in idiopathic pulmonary fibrosis and their relationship to disease extent. Am J Respir Crit Care Med. 2000;162:1109–14.PubMedCrossRef
8.
Zurück zum Zitat Ohnishi H, Yokoyama A, Kondo K, Hamada H, Abe M, Nishimura K, et al. Comparative study of KL-6, surfactant protein-a, surfactant protein-D, and monocyte chemoattractant protein-1 as serum markers for interstitial lung diseases. Am J Respir Crit Care Med. 2002;165:378–81.PubMedCrossRef Ohnishi H, Yokoyama A, Kondo K, Hamada H, Abe M, Nishimura K, et al. Comparative study of KL-6, surfactant protein-a, surfactant protein-D, and monocyte chemoattractant protein-1 as serum markers for interstitial lung diseases. Am J Respir Crit Care Med. 2002;165:378–81.PubMedCrossRef
9.
Zurück zum Zitat Ohta S, Okamoto M, Fujimoto K, Sakamoto N, Takahashi K, Yamamoto H, et al. The usefulness of monomeric periostin as a biomarker for idiopathic pulmonary fibrosis. PLoS One. 2017;12:e0174547.PubMedPubMedCentralCrossRef Ohta S, Okamoto M, Fujimoto K, Sakamoto N, Takahashi K, Yamamoto H, et al. The usefulness of monomeric periostin as a biomarker for idiopathic pulmonary fibrosis. PLoS One. 2017;12:e0174547.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Kakugawa T, Yokota S, Ishimatsu Y, Hayashi T, Nakashima S, Hara S, et al. Serum heat shock protein 47 levels are elevated in acute exacerbation of idiopathic pulmonary fibrosis. Cell Stress Chaperones. 2013;18:581–90.PubMedPubMedCentralCrossRef Kakugawa T, Yokota S, Ishimatsu Y, Hayashi T, Nakashima S, Hara S, et al. Serum heat shock protein 47 levels are elevated in acute exacerbation of idiopathic pulmonary fibrosis. Cell Stress Chaperones. 2013;18:581–90.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Rhee SG, Chae HZ, Kim K. Peroxiredoxins: a historical overview and speculative preview of novel mechanisms and emerging concepts in cell signaling. Free Radic Biol Med. 2015;38:1543–52.CrossRef Rhee SG, Chae HZ, Kim K. Peroxiredoxins: a historical overview and speculative preview of novel mechanisms and emerging concepts in cell signaling. Free Radic Biol Med. 2015;38:1543–52.CrossRef
12.
Zurück zum Zitat Kang SW, Rhee SG, Chang TS, Jeong W, Choi MH. 2-Cys peroxiredoxin function in intracellular signal transduction: therapeutic implications. Trends Mol Med. 2005;11:571–8.PubMedCrossRefPubMedCentral Kang SW, Rhee SG, Chang TS, Jeong W, Choi MH. 2-Cys peroxiredoxin function in intracellular signal transduction: therapeutic implications. Trends Mol Med. 2005;11:571–8.PubMedCrossRefPubMedCentral
13.
Zurück zum Zitat Jin DY, Chae HZ, Rhee SG, Jeang KT. Regulatory role for a novel human thioredoxin peroxidase in NF-kappaB activation. J Biol Chem. 1997;272:30952–61.PubMedCrossRef Jin DY, Chae HZ, Rhee SG, Jeang KT. Regulatory role for a novel human thioredoxin peroxidase in NF-kappaB activation. J Biol Chem. 1997;272:30952–61.PubMedCrossRef
14.
Zurück zum Zitat Giguere P, Turcotte ME, Hamelin E, Parent A, Brisson J, Laroche G, et al. Peroxiredoxin-4 interacts with and regulates the thromboxane a(2) receptor. FEBS Lett. 2007;581:3863–8.PubMedCrossRef Giguere P, Turcotte ME, Hamelin E, Parent A, Brisson J, Laroche G, et al. Peroxiredoxin-4 interacts with and regulates the thromboxane a(2) receptor. FEBS Lett. 2007;581:3863–8.PubMedCrossRef
15.
Zurück zum Zitat Matsumoto A, Okado A, Fujii T, Fujii J, Egashira M, Niikawa N, et al. Cloning of the peroxiredoxin gene family in rats and characterization of the fourth member. FEBS Lett. 1999;443:246–50.PubMedCrossRef Matsumoto A, Okado A, Fujii T, Fujii J, Egashira M, Niikawa N, et al. Cloning of the peroxiredoxin gene family in rats and characterization of the fourth member. FEBS Lett. 1999;443:246–50.PubMedCrossRef
16.
Zurück zum Zitat Kikuchi N, Ishii Y, Morishima Y, Yageta Y, Haraguchi N, Yamadori T, et al. Aggravation of bleomycin-induced pulmonary inflammation and fibrosis in mice lacking peroxiredoxin I. Am J Respir Cell Mol Biol. 2011;45:600–9.PubMedCrossRef Kikuchi N, Ishii Y, Morishima Y, Yageta Y, Haraguchi N, Yamadori T, et al. Aggravation of bleomycin-induced pulmonary inflammation and fibrosis in mice lacking peroxiredoxin I. Am J Respir Cell Mol Biol. 2011;45:600–9.PubMedCrossRef
17.
Zurück zum Zitat Vuorinen K, Ohlmeier S, Leppäranta O, Salmenkivi K, Myllärniemi M, Kinnula VL. Peroxiredoxin II expression and its association with oxidative stress and cell proliferation in human idiopathic pulmonary fibrosis. J Histochem Cytochem. 2008;56:951–9.PubMedPubMedCentralCrossRef Vuorinen K, Ohlmeier S, Leppäranta O, Salmenkivi K, Myllärniemi M, Kinnula VL. Peroxiredoxin II expression and its association with oxidative stress and cell proliferation in human idiopathic pulmonary fibrosis. J Histochem Cytochem. 2008;56:951–9.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Guo X, Yamada S, Tanimoto A, Ding Y, Wang KY, Shimajiri S, et al. Overexpression of peroxiredoxin 4 attenuates atherosclerosis in apolipoprotein E knockout mice. Antioxid Redox Signal. 2012;17:1362–75.PubMedPubMedCentralCrossRef Guo X, Yamada S, Tanimoto A, Ding Y, Wang KY, Shimajiri S, et al. Overexpression of peroxiredoxin 4 attenuates atherosclerosis in apolipoprotein E knockout mice. Antioxid Redox Signal. 2012;17:1362–75.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Nawata A, Noguchi H, Mazaki Y, Kurahashi T, Izumi H, Wang KY, et al. Overexpression of peroxiredoxin 4 affects intestinal function in a dietary mouse model of nonalcoholic fatty liver disease. PLoS One. 2016;11:e0152549.PubMedPubMedCentralCrossRef Nawata A, Noguchi H, Mazaki Y, Kurahashi T, Izumi H, Wang KY, et al. Overexpression of peroxiredoxin 4 affects intestinal function in a dietary mouse model of nonalcoholic fatty liver disease. PLoS One. 2016;11:e0152549.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Collard HR, Moore BB, Flaherty KR, Brown KK, Kaner RJ, King TE Jr, et al. Acute exacerbations of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2007;176:636–43.PubMedCrossRef Collard HR, Moore BB, Flaherty KR, Brown KK, Kaner RJ, King TE Jr, et al. Acute exacerbations of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2007;176:636–43.PubMedCrossRef
22.
Zurück zum Zitat Travis WD, Costabel U, Hansell DM, King TE Jr, Lynch DA, Nicholson AG, et al. An official American Thoracic Society/European Respiratory Society statement: update of the international multidisciplinary classification of the idiopathic interstitial pneumonias. Am J Respir Crit Care Med. 2013;188:733–48.PubMedPubMedCentralCrossRef Travis WD, Costabel U, Hansell DM, King TE Jr, Lynch DA, Nicholson AG, et al. An official American Thoracic Society/European Respiratory Society statement: update of the international multidisciplinary classification of the idiopathic interstitial pneumonias. Am J Respir Crit Care Med. 2013;188:733–48.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Ding Y, Yamada S, Wang KY, Shimajiri S, Guo X, Tanimoto A, et al. Overexpression of peroxiredoxin 4 protects against high-dose streptozotocin-induced diabetes by suppressing oxidative stress and cytokines in transgenic mice. Antioxid Redox Signal. 2010;13:1477–90.PubMedCrossRef Ding Y, Yamada S, Wang KY, Shimajiri S, Guo X, Tanimoto A, et al. Overexpression of peroxiredoxin 4 protects against high-dose streptozotocin-induced diabetes by suppressing oxidative stress and cytokines in transgenic mice. Antioxid Redox Signal. 2010;13:1477–90.PubMedCrossRef
24.
Zurück zum Zitat Ashcroft T, Simpson JM, Timbrell V. Simple method of estimating severity of pulmonary fibrosis on a numerical scale. J Clin Pathol. 1988;41:467–70.PubMedPubMedCentralCrossRef Ashcroft T, Simpson JM, Timbrell V. Simple method of estimating severity of pulmonary fibrosis on a numerical scale. J Clin Pathol. 1988;41:467–70.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Nabeshima A, Yamada S, Guo X, Tanimoto A, Wang KY, Shimajiri S, et al. Peroxiredoxin 4 protects against nonalcoholic steatohepatitis and type 2 diabetes in a nongenetic mouse model. Antioxid Redox Signal. 2013;19:1983–98.PubMedPubMedCentralCrossRef Nabeshima A, Yamada S, Guo X, Tanimoto A, Wang KY, Shimajiri S, et al. Peroxiredoxin 4 protects against nonalcoholic steatohepatitis and type 2 diabetes in a nongenetic mouse model. Antioxid Redox Signal. 2013;19:1983–98.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Noguchi S, Yatera K, Wang KY, Oda K, Akata K, Yamasaki K, et al. Nitric oxide exerts protective effects against bleomycin-induced pulmonary fibrosis in mice. Respir Res. 2014;15:92.PubMedPubMedCentralCrossRef Noguchi S, Yatera K, Wang KY, Oda K, Akata K, Yamasaki K, et al. Nitric oxide exerts protective effects against bleomycin-induced pulmonary fibrosis in mice. Respir Res. 2014;15:92.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Noguchi H, Yamada S, Nabeshima A, Guo X, Tanimoto A, Wang KY, et al. Depletion of apoptosis signal-regulating kinase 1 prevents bile duct ligation-induced necroinflammation and subsequent peribiliary fibrosis. Am J Pathol. 2014;184:644–61.PubMedCrossRef Noguchi H, Yamada S, Nabeshima A, Guo X, Tanimoto A, Wang KY, et al. Depletion of apoptosis signal-regulating kinase 1 prevents bile duct ligation-induced necroinflammation and subsequent peribiliary fibrosis. Am J Pathol. 2014;184:644–61.PubMedCrossRef
28.
Zurück zum Zitat Sakamoto N, Ishimatsu Y, Kakugawa T, Yura H, Tomonaga M, Harada T, et al. Elevated plasma alpha-defensins in patients with acute exacerbation of fibrotic interstitial pneumonia. Respir Med. 2015;109:265–71.PubMedCrossRef Sakamoto N, Ishimatsu Y, Kakugawa T, Yura H, Tomonaga M, Harada T, et al. Elevated plasma alpha-defensins in patients with acute exacerbation of fibrotic interstitial pneumonia. Respir Med. 2015;109:265–71.PubMedCrossRef
29.
Zurück zum Zitat Ishizaka A, Matsuda T, Albertine KH, Koh H, Tasaka S, Hasegawa N, et al. Elevation of KL-6, a lung epithelial cell marker, in plasma and epithelial lining fluid in acute respiratory distress syndrome. Am J Physiol Lung Cell Mol Physiol. 2004;286:L1088–94.PubMedCrossRef Ishizaka A, Matsuda T, Albertine KH, Koh H, Tasaka S, Hasegawa N, et al. Elevation of KL-6, a lung epithelial cell marker, in plasma and epithelial lining fluid in acute respiratory distress syndrome. Am J Physiol Lung Cell Mol Physiol. 2004;286:L1088–94.PubMedCrossRef
30.
Zurück zum Zitat Enomoto Y, Matsushima S, Shibata K, Aoshima Y, Yagi H, Meguro S, et al. LTBP2 is secreted from lung myofibroblasts and is a potential biomarker for idiopathic pulmonary fibrosis. Clin Sci (Lond). 2018;132:1565–80.CrossRef Enomoto Y, Matsushima S, Shibata K, Aoshima Y, Yagi H, Meguro S, et al. LTBP2 is secreted from lung myofibroblasts and is a potential biomarker for idiopathic pulmonary fibrosis. Clin Sci (Lond). 2018;132:1565–80.CrossRef
31.
Zurück zum Zitat Hirasawa Y, Kohno N, Yokoyama A, Inoue Y, Abe M, Hiwada K. KL-6, a human MUC1 mucin, is chemotactic for human fibroblasts. Am J Respir Cell Mol Biol. 1997;17:501–7.PubMedCrossRef Hirasawa Y, Kohno N, Yokoyama A, Inoue Y, Abe M, Hiwada K. KL-6, a human MUC1 mucin, is chemotactic for human fibroblasts. Am J Respir Cell Mol Biol. 1997;17:501–7.PubMedCrossRef
32.
Zurück zum Zitat Nickel CH, Ruedinger J, Misch F, Blume K, Maile S, Schulte J, et al. Copeptin and peroxiredoxin-4 independently predict mortality in patients with nonspecific complaints presenting to the emergency department. Acad Emerg Med. 2011;18:851–9.PubMedCrossRef Nickel CH, Ruedinger J, Misch F, Blume K, Maile S, Schulte J, et al. Copeptin and peroxiredoxin-4 independently predict mortality in patients with nonspecific complaints presenting to the emergency department. Acad Emerg Med. 2011;18:851–9.PubMedCrossRef
33.
Zurück zum Zitat Schulte J, Struck J, Kohrle J, Muller B. Circulating levels of peroxiredoxin 4 as a novel biomarker of oxidative stress in patients with sepsis. Shock. 2011;35:460–5.PubMedCrossRef Schulte J, Struck J, Kohrle J, Muller B. Circulating levels of peroxiredoxin 4 as a novel biomarker of oxidative stress in patients with sepsis. Shock. 2011;35:460–5.PubMedCrossRef
34.
Zurück zum Zitat Oku H, Shimizu T, Kawabata T, Nagira M, Hikita I, Ueyama A, et al. Antifibrotic action of pirfenidone and prednisolone: different effects on pulmonary cytokines and growth factors in bleomycin-induced murine pulmonary fibrosis. Eur J Pharmacol. 2008;590:400–8.PubMedCrossRef Oku H, Shimizu T, Kawabata T, Nagira M, Hikita I, Ueyama A, et al. Antifibrotic action of pirfenidone and prednisolone: different effects on pulmonary cytokines and growth factors in bleomycin-induced murine pulmonary fibrosis. Eur J Pharmacol. 2008;590:400–8.PubMedCrossRef
35.
Zurück zum Zitat Wilson MS, Madala SK, Ramalingam TR, Gochuico BR, Rosas IO, Cheever AW, et al. Bleomycin and IL-1beta-mediated pulmonary fibrosis is IL-17A dependent. J Exp Med. 2010;207:535–52.PubMedPubMedCentralCrossRef Wilson MS, Madala SK, Ramalingam TR, Gochuico BR, Rosas IO, Cheever AW, et al. Bleomycin and IL-1beta-mediated pulmonary fibrosis is IL-17A dependent. J Exp Med. 2010;207:535–52.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Gasse P, Riteau N, Vacher R, Michel ML, Fautrel A, di Padova F, et al. IL-1 and IL-23 mediate early IL-17A production in pulmonary inflammation leading to late fibrosis. PLoS One. 2011;6:e23185.PubMedPubMedCentralCrossRef Gasse P, Riteau N, Vacher R, Michel ML, Fautrel A, di Padova F, et al. IL-1 and IL-23 mediate early IL-17A production in pulmonary inflammation leading to late fibrosis. PLoS One. 2011;6:e23185.PubMedPubMedCentralCrossRef
37.
38.
Zurück zum Zitat Shichita T, Hasegawa E, Kimura A, Morita R, Sakaguchi R, Takada I, et al. Peroxiredoxin family proteins are key initiators of post-ischemic inflammation in the brain. Nat Med. 2012;18:911–7.PubMedCrossRef Shichita T, Hasegawa E, Kimura A, Morita R, Sakaguchi R, Takada I, et al. Peroxiredoxin family proteins are key initiators of post-ischemic inflammation in the brain. Nat Med. 2012;18:911–7.PubMedCrossRef
39.
Zurück zum Zitat Iwakura Y, Ishigame H, Saijo S, Nakae S. Functional specialization of interleukin-17 family members. Immun. 2011;34:149–62.CrossRef Iwakura Y, Ishigame H, Saijo S, Nakae S. Functional specialization of interleukin-17 family members. Immun. 2011;34:149–62.CrossRef
40.
Zurück zum Zitat Kinnula VL, Fattman CL, Tan RJ, Oury TD. Oxidative stress in pulmonary fibrosis: a possible role for redox modulatory therapy. Am J Respir Crit Care Med. 2005;172:417–22.PubMedPubMedCentralCrossRef Kinnula VL, Fattman CL, Tan RJ, Oury TD. Oxidative stress in pulmonary fibrosis: a possible role for redox modulatory therapy. Am J Respir Crit Care Med. 2005;172:417–22.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Kuwano K, Nakashima N, Inoshima I, Hagimoto N, Fujita M, Yoshimi M, et al. Oxidative stress in lung epithelial cells from patients with idiopathic interstitial pneumonias. Eur Respir J. 2003;21:232–40.PubMedCrossRef Kuwano K, Nakashima N, Inoshima I, Hagimoto N, Fujita M, Yoshimi M, et al. Oxidative stress in lung epithelial cells from patients with idiopathic interstitial pneumonias. Eur Respir J. 2003;21:232–40.PubMedCrossRef
42.
Zurück zum Zitat Wang Y, Feinstein SI, Manevich Y, Ho YS, Fisher AB. Peroxiredoxin 6 gene-targeted mice show increased lung injury with paraquat-induced oxidative stress. Antioxid Redox Signal. 2006;8:229–37.PubMedCrossRef Wang Y, Feinstein SI, Manevich Y, Ho YS, Fisher AB. Peroxiredoxin 6 gene-targeted mice show increased lung injury with paraquat-induced oxidative stress. Antioxid Redox Signal. 2006;8:229–37.PubMedCrossRef
43.
Zurück zum Zitat Papiris SA, Tomos IP, Karakatsani A, Spathis A, Korbila I, Analitis A, et al. High levels of IL-6 and IL-8 characterize early-on idiopathic pulmonary fibrosis acute exacerbations. Cytokine. 2018;102:168–72.PubMedCrossRef Papiris SA, Tomos IP, Karakatsani A, Spathis A, Korbila I, Analitis A, et al. High levels of IL-6 and IL-8 characterize early-on idiopathic pulmonary fibrosis acute exacerbations. Cytokine. 2018;102:168–72.PubMedCrossRef
Metadaten
Titel
The overexpression of peroxiredoxin-4 affects the progression of idiopathic pulmonary fibrosis
verfasst von
Tetsuya Hanaka
Takashi Kido
Shingo Noguchi
Sohsuke Yamada
Hirotsugu Noguchi
Xin Guo
Aya Nawata
Ke-Yong Wang
Keishi Oda
Tsutomu Takaki
Hiroto Izumi
Hiroshi Ishimoto
Kazuhiro Yatera
Hiroshi Mukae
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
BMC Pulmonary Medicine / Ausgabe 1/2019
Elektronische ISSN: 1471-2466
DOI
https://doi.org/10.1186/s12890-019-1032-2

Weitere Artikel der Ausgabe 1/2019

BMC Pulmonary Medicine 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.