Skip to main content
Erschienen in: BMC Pulmonary Medicine 1/2018

Open Access 01.12.2018 | Research article

Annexin A1 in plasma from patients with bronchial asthma: its association with lung function

verfasst von: Sun-Hye Lee, Pureun-Haneul Lee, Byeong-Gon Kim, Hyun-Jeong Seo, Ae-Rin Baek, Jong-Sook Park, June-Hyuk Lee, Sung-Woo Park, Do-Jin Kim, Choon-Sik Park, An-Soo Jang

Erschienen in: BMC Pulmonary Medicine | Ausgabe 1/2018

Abstract

Background

Annexin-A1 (ANXA1) is a glucocorticoid-induced protein with multiple actions in the regulation of inflammatory cell activation. The anti-inflammatory protein ANXA1 and its N-formyl peptide receptor 2 (FPR2) have protective effects on organ fibrosis. However, the exact role of ANXA1 in asthma remains to be determined. The aim of this study was to identify the role of ANXA1 in bronchial asthma.

Methods

In mice sensitized and challenged with ovalbumin (OVA-OVA mice) and mice sensitized with saline and challenged with air (control mice), we investigated the potential links between ANXA1 levels and bronchial asthma using ELISA, immunoblotting, and immunohistochemical staining. Moreover, we also determined ANXA1 levels in blood from 50 asthmatic patients (stable and exacerbated states).

Results

ANXA1 protein levels in lung tissue and bronchoalveolar lavage fluid were significantly higher in OVA-OVA mice compared with control mice. FPR2 protein levels in lung tissue were significantly higher in OVA-OVA mice compared with control mice. Plasma ANXA1 levels were increased in asthmatic patients compared with healthy controls. Plasma ANXA1 levels were significantly lower in exacerbated patients compared with stable patients with bronchial asthma (p < 0.05). The plasma ANXA1 levels in controlled asthmatic patients were correlated with forced expiratory volume in 1 s (FEV1) (r = − 0.191, p = 0.033) and FEV1/forced vital capacity (FVC) (r = −0.202, p = 0.024).

Conclusion

These results suggest that ANXA1 may be a potential marker and therapeutic target for asthma.
Abkürzungen
AHR
Airway hyperresponsiveness
ANXA1
Annexin A1
AUC
Area under curve
BA
Bronchial asthma
BALF
Bronchoalveolar lavage fluid
DEX
Dexamethasone
ELISA
Enzyme-linked immunosorbent assay
FEV1
Forced expiratory volume in the first second
FPR2
N-formyl peptide receptor 2
FVC
Forced vital capacity
IQR
Interquartile range
LXA4
Lipoxin A4
OVA
Ovalbumin
ROC
Receiver operating characteristic curve
TiO2
Titanium dioxide

Background

Annexin A1 (ANXA1) is a glucocorticoid-induced protein with multiple functions in the regulation of inflammatory cell activation. ANXA1 is a 37 kDa protein that can bind to cellular membranes in a Ca2 + −dependent manner. The protein has been reported to have anti-phospholipase activity following glucocorticoid induction and possesses a wide range of physiological and pathological functions [16]. The biological effects of ANXA1 differ based on intra- versus extracellular localization [79]. The extracellular form of ANXA1 stimulates cell motility and cancer cell invasion, mostly via interaction with specific receptors such as the G-protein-coupled formyl peptide receptor (FPR) family [3, 10, 11].
ANXA1 has anti-inflammatory effects by stimulating inflammatory cell programmed cell death and prohibiting eicosanoid synthesis [12, 13]. ANXA1 levels were decreased in smokers or patients with asthma, cystic fibrosis, and rheumatoid arthritis [1417]. The reduced levels of lipoxin A4 (LXA4) and ANXA1 were reported in wheezy infants [17] and patients with severe asthma [1820]. In addition, experimental studies have shown that ANXA1 is associated with asthma development [16].
However, the role of ANXA1 in the pathogenesis of asthma is not clear. Using a mouse model, we evaluated ANXA1 expression and levels in the blood of asthmatic patients and evaluated the relationship between ANXA1 levels and clinical profiles in asthma.

Methods

Experimental design

Fifty asthmatic patients were recruited and followed for 6.6 ± 3.6 years, and plasma ANXA1 levels were determined during the stable and exacerbated states (Table 1). Experiments involving patients were approved by Soonchunhyang University’s institutional review board (IRB). Eight BALB/c mice were exposed to saline (control) or ovalbumin (OVA). Detailed analyses included evaluation of lung ANXA1 phospho Tyr21 levels and lung histology and FPR2 levels in bronchoalveolar lavage fluid (BALF) and lung tissue. Cells were treated with titanium dioxide nanoparticles (TiO2) and dexamethasone (DEX). The animal studies were approved by Soonchunhyang University’s institutional animal care and use committee.
Table 1
Clinical characteristics in control subjects and patients with asthma
Characteristic
Control subjects
Asthmatic patients
Stable
Exacerbated
No of subjects
25
50
Sex (male/female)
2/23
20/30
Age (of initial visit), yr
58.3 ± 6.2
54.9 ± 14.1
Onset of asthma: age, yr
 
47.06 ± 17.25
Asthma duration, yr
 
6.63 ± 3.60
Smoking status (NS/ES/CS)
25/0/0
32/12/6
Cigarettes smoked, pack. yr
 
9.0 ± 15.7
Body Mass Index, kg/ m2
24.8 ± 2.61
25.4 ± 3.31
Stable and exacerbate lung function
FEV1, % pred.
115.36 ± 16.59
85.43 ± 19.82
62.60 ± 18.14
FVC, % pred.
96.56 ± 14.51
84.83 ± 16.68
66.51 ± 16.76
FEV1 /FVC
84.24 ± 6.05
74.53 ± 9.72
68.07 ± 11.84
PC20, mg/ml
 
9.18 ± 10.37
Total IgE, kU
106.63 ± 188.7
421.5 ± 699.19*
Atopy
1 (4%)
21 (42%)*
Attack average/yr
 
3.38 ± 3.24
Duration of exacerbation during follow up
 
6.63 ± 3.61
Blood WBC/uL
5587.2 ± 1268.0
7768.8 ± 3448.9*
9873.0 ± 4917.1
Blood eosinophil, %
2.73 ± 2.26
5.09 ± 4.97*
3.83 ± 5.42
Blood neutrophil, %
56.25 ± 9.8
54.7 ± 12.9
64.6 ± 19.3
Data expressed as mean ± SD. BMI body mass index, ES ex-smoker, FEV 1 forced expiratory volume in one second, FVC forced vital capacity, NS non-smoker, PC20 methacholine; the concentration of methacholine required to decrease the FEV1 by 20%, SM; smoker. *p < 0.01 compared with control subjects, p < 0.05 compared with stable asthmatics

Subjects

All patients were recruited from Soonchunhyang University, Bucheon Hospital. Asthma diagnoses were based on the Global Initiative for Asthma (GINA) guidelines [21]. This study used the same sample examined by Moon et al. [22] although several different measures and analyses are presented. The biospecimens and data used in this study were provided by the biobank of Soonchunhyang University Bucheon Hospital, a member of the Korean Biobank Network.
All subjects had a clinical diagnosis of asthma supported by one or more of the following criteria: 1) variation in the maximum diurnal peak expiratory flow >20% over the course of 14 days, 2) an increase in forced expiratory volume in 1 s (FEV1) of >15% after inhalation of 200–400 μg albuterol, or 3) a 20% reduction in FEV1 in response to a stimulating concentration of inhaled methacholine (PC20 methacholine) < 10 mg/ml. All subjects underwent standardized assessments, which included analyses of induced sputum specimens, complete differential blood cell counts, immunoglobulin E (IgE) measurements, chest posteroanterior radiography, allergy skin prick tests, and spirometry. All data were collected at the time of diagnosis, before administration of the asthma medication.
Among asthmatic patients from the hospital, asthmatics matched to normal controls in terms of age, sex, and BMI were selected for the study. Normal control subjects were recruited from among the spouses of the patients or members of the general population who answered negatively to a screening questionnaire regarding respiratory symptoms and other allergic diseases, had FEV1 values >80% predicted, PC20 methacholine level > 10 mg/ml, and normal findings on simple chest radiographs. Of the subjects who completed a follow-up period of at least 2 years, 50 were diagnosed using the GINA guidelines [21].

Asthma exacerbation

Asthma exacerbation was analyzed in subjects who had completed regular follow-up for at least 2 years. Asthma exacerbation was defined by the GINA guidelines as episodes of progressively increasing shortness of breath, cough, wheezing, chest tightness, or some combination of these symptoms, accompanied by decreased expiratory airflow and use of systemic corticosteroids (tablets, suspensions, or injections) or an increase in dose from the stable maintenance dose for at least 3 days and a hospitalization or emergency department visit due to asthma, requiring systemic corticosteroids.

Elisa

ANXA1 levels in the blood of asthmatic patients was measured by Enzyme-linked immunosorbent assay (ELISA; R&D System, Minneapolis, MN, USA). To compare results from different plates, the optical densities (ODs) of the test samples were adjusted relative to the positive and negative control samples supplied in each kit. The mean OD of duplicate wells was calculated. The index value of each test sample was defined by the following formula: index = (OD of test sample – OD of negative control)/(OD of positive control – OD of negative control) × 100. Low detection limits were set at 0.119 ng/mL for ANXA1 according to the manufacturer’s recommendation.

Animals

Female 6-week-old BALB/c mice (6 weeks of age, weighing 20–24 g) were purchased from Charles River Korea (Orient Bio Inc., Seongnam, Korea). All mice were sensitized by intraperitoneal injection on days 0 and 14 with 50 μg grade V chicken egg OVA (Sigma-Aldrich, St Louis, MO, USA) emulsified with 10 mg hydroxyl aluminum in 100 μl Dulbecco’s phosphate buffered saline (D-PBS). On days 21–23, all mice received intranasal challenges with 150 μg grade III OVA (Sigma-Aldrich) in 50 μl D-PBS. Control mice were sensitized and challenged with saline. On day 24, airway hyper responsiveness (AHR) was measured, BALF was collected, and lung tissue was processed for protein, RNA, and hematoxylin and eosin (H&E) staining and immunohistochemistry (IHC).

AHR, BALF, and morphology analysis

Mice were anesthetized with 2.5 mg/kg tiletamine and xylazine (Zoletil and lumpum; Bayer Korea Co, Seoul, Korea), and AHR was assessed following challenges with 0, 5, 20, or 100 mg/ml methacholine (Sigma-Aldrich). Measurements of airway hyperresponsiveness were conducted using an animal pulmonary instrument (OCP-3000) 1 min after each dose with 3 min between doses. The following day, BALF was obtained, centrifuged, and the supernatant stored (−20 °C). The cell pellet was resuspended for cell counting, and cytospin slides were prepared for stained with modified Diff-Quick stain. Differential cell counting was performed on at least 500 cells in each slide using standard morphological criteria under a light microscope. A portion of the lung was fixed in 4% phosphate-buffered paraformaldehyde, embedded in paraffin, sectioned (4 μm), and stained (H&E and IHC staining).

Immunohistochemistry

Mouse lung sections were deparaffinized and rehydrated in an ethanol gradient series. The sections were incubated with 1.4% hydrogen peroxide in methanol for 30 min to block endogenous peroxidases, with 1.5% horse serum to block non-specific binding, and then with the anti-rabbit ANXA1 primary antibody (1200, Thermo Fisher, Rockford, IL, USA). The next day, sections were treated with the ABC kit (Vector Laboratories, Burlingame, CA, USA). The color reaction was developed by staining with liquid DAB+ substrate (Golden Bridge International Inc., Mukilteo, WA, USA). After immunohistochemical staining, the slides were counterstained with Harris’s hematoxylin for 1 min. Images were analyzed with the ImageJ program (National Institutes of Health, Bethesda, Md).

Western blot

The extracted lung tissues were homogenized in a protein lysis solution containing 50 mM Tris-HCl (pH 7.4), 50 mM NaCl, 0.1% SDS, 1% Triton X-100, 0.5 mM EDTA, and 100 mM PMSF in distilled water. After centrifuged at 14,000 rpm for 30 min at 4 °C, the soluble materials were collected. Mouse lung lysates and bronchoalveolar lavage fluid (BALF) proteins were separated by SDS-PAGE and transferred to PVDF membranes. The membranes were blocked in TBS with 5% skim milk and 0.1% Tween 20 for 1 h at room temperature before incubating with rabbit anti-ANXA1 (1:1000, Thermo Fisher) or rabbit anti-FPRL1/FPR2 (Novus Biologicals, Littleton, USA) or rabbit anti-ANXA1 (phospho Tyr21) (1:500, Genetex, San Antonio, TX, USA) (overnight, 4 °C). The membranes were then incubated for 1 h at room temperature with an HRP-conjugated secondary antibody (1:5000, Santa Cruz Biotechnology, Dallas, USA). Detection was performed using an enhanced chemiluminescence (ECL) plus Western Blot Detection System (ATTO, Tokyo, Japan) on X-ray film. The relative protein levels were determined by quantitative densitometry and were normalized to anti-β-actin monoclonal antibody (1:5000, Sigma-Aldrich) levels.

Cell culture

Normal human bronchial primary epithelial cells (NHBE) were purchased from Lonza (Lonza, Basel, Switzerland, cat#. CC-2540). NHBE cells were plated at 3000 cells/cm2 in culture 75 cm2 flasks in bronchial epithelial cell growth medium supplemented with the BEGM BulletKit™ (Bronchial Epithelial Cell Growth Medium) (Lonza, cat#: CC-3170) and cultured at 37 °C in a 5% CO2 incubator. The medium was changed every 48 h, and the cells were grown to 80–90% confluence for 5 to 6 days. NHBE cells (second-passage) with density 1.5 × 106 cells/ml were seeded on in 6-well plate with BEGM medium. The 24 h prior to the experiment, the medium was changed to BEBM basal medium. The cells were exposed to 100 μM TiO2 and 10 μM DEX (Sigma-Aldrich, Melbourne, VIC, Australia). Additionally, control cell lines are not exposed to DEX and TiO2. Cells were treated with different doses time (4, 8, 24 h).

Statistical analysis

Receiver operating characteristic (ROC) curve, the area under the curve (AUC) was calculated using significant predictors (as determined via multivariate regression) to derive best suitable cut-off values and to assess model discrimination and predictive accuracy. Determination of optimal cutoff points were conducted with SPSS statistical software package (ver. 20.0; SPSS Inc.; Chicago, IL, USA).
The data were double-entered into the SPSS statistical software package (20).
Data with a normal distribution were described by means and standard deviations and compared using a two-sample t-test; data with non-parametric distributions were described by the median and interquartile ranges (IQR) and compared using Mann-Whitney U-test (intergroup analysis) or Pearson’s χ2 test for normally distributed, skewed, and categorical data, respectively. Differences between the patient populations were analyzed by χ2 test with Fisher’s exact test when low expected cell counts were encountered. Different correlation coefficients were calculated the Spearman’s correlation analysis. p < 0.05 was deemed to indicate statistical significance.

Results

ANXA1 plasma levels in asthmatic patients

Plasma ANXA1 levels in asthmatic patients (0.570 ± 0.043 ng/ml) were higher than those in healthy control subjects (0.312 ± 0.078 ng/ml; Interquartile range, IQR: 0.0–0.611), (p < 0.01) (Fig. 1a). Plasma ANXA1 levels were significantly lower in exacerbated patients (0.497 ± 0.058 ng/ml; IQR: 0.038–0.850) compared with stable patients (0.645 ± 0.061 ng/ml; IQR: 0.447–0.871), (p < 0.05) (Fig. 1a). Therefore, this study calculated the best cut-off values of the Annexin-A1 ELISA in patients. By receiver operating characteristic curve (ROC), the best cut-off value of Annexin-A1 ELISA was 0.5023 ng/mL in patients (sensitivity and specificity were 68% and 99.68%, resp.), and the area under curve (AUC) was 0.680 (95% CI; 0.596–0.829), (p < 0.001) (Fig. 1b).

Relationship between ANXA1 levels and clinical variables

The population characteristics are shown in Table 1. Plasma ANXA1 levels in controlled asthmatic patients were correlated with forced expiratory volume in 1 s (FEV1%) pred. (r    = −0.191, p = 0.033) (Fig. 2a) and FEV1/forced vital capacity (FVC), (r = −0.202, p = 0.024) (Fig. 2b). Smoking asthmatics tended to have lower levels of ANXA1 compared with non-smoking asthmatics (4.14 ± 0.31 vs. 4.56 ± 0.99 ng/ml; p > 0.05). There was no relationship between ANXA1 and age, smoking amount, body mass index, PC20, white blood cell count, or total IgE levels.

Airway hyperresponsiveness and differential cell counts in mouse asthma model

Airway hyperresponsiveness (AHR) was increased in the OVA sensitized and OVA challenged mice (OVA-OVA mice group) compared with saline sensitized and air challenged mice (Control mice group) (Fig. 3a). Inflammatory cells, including eosinophils, macrophages, lymphocytes and neutrophils were increased in the BAL fluid in OVA-OVA mice compared with control mice group (Fig. 3b). The results demonstrated that induced the infiltration of inflammatory cells into the BAL fluid of the OVA-OVA challenged mice.

ANXA1 in a mouse model of asthma and NHBE cells

ANXA1 protein levels were significantly increased in lung tissue (Fig. 4) and bronchoalveolar lavage fluid (BALF) (Fig. 5) of mice sensitized and challenged with OVA (OVA-OVA mice) compared with mice sensitized with saline and challenged with air (control mice). FPR2 (Fig. 6) protein levels were significantly increased in the lung tissue of OVA-OVA mice compared with control mice. ANXA1-phospho Tyr21 protein levels were significantly increased in the lung tissue (Fig. 7) of OVA-OVA mice compared with control mice. ANXA1 protein levels were increased (Fig. 8) in dexamethasone (DEX) and titanium dioxide (TiO2) treated NHBE cells at 4, 8, 24 h (p < 0.05, controls vs. DEX or DEX + TiO2 or TiO2).

Discussion

In this study, plasma ANXA1 levels were increased in patients with asthma but decreased in patients with exacerbated asthma. ANXA1 levels were also correlated with lung function, suggesting that ANXA1 may be a potential marker for asthma.
Annexins are soluble proteins that bind to cell membranes containing negatively charged phospholipids, principally phosphatidylserine, in a Ca2 + −dependent manner. The contribution of anti-inflammatory protein ANXA1 and its receptor FPR2 to the regulation of inflammatory responses in human normal lung fibroblasts has been reported previously [2325]. ANXA1 and FPR2 have protective effects in organ fibrosis [26].
ANXA1, an abundant intracellular protein expressed in many cell types, has known to be induced by glucocorticoids (GCs) and to inhibit phospholipase activity [2429]. Recombinant ANXA1 or ANXA1-derived N-terminal peptides has similar actions like the anti-inflammatory action of glucocorticoids such as inhibition of inflammatory cells, and suppressing inflammatory mediators [26, 30].
In this study, total and phosphorylated ANXA1 levels were increased in a mouse model of asthma and in inflammatory, endothelial, and epithelial cells. In addition, receptor FPR2 expression was increased in OVA-challenged mice, suggesting that ANXA1 may be involved in asthma pathogenesis.
The ANXA1 receptor FPR2 as a specific G-protein-coupled receptor binds to LXA4 (an anti-inflammatory lipid) and serum amyloid protein, which mediate ligand-specific effects [30, 31]. FPR2 has expressed in human lung fibroblasts, and induced by GCs in human myeloid cells [32, 33].
ANXA1 levels in bronchoalveolar lavage fluids were higher in smokers [14] and patients with cystic fibrosis [15]. A form of ANXA1 with a molecular weight of 33 kDa is released rather than the 37 kDa ANXA1, suggesting that ANXA1 be degraded in smokers and patients with cystic fibrosis [16, 17]. Ng et al. [16] reported that an ANXA-1-deficient mice exhibit spontaneous airway hyperresponsiveness and exacerbated allergen-specific antibody responses in a mouse model of asthma.
In this study plasma levels of ANXA1 were increased in asthmatic patients compared with healthy control subjects, which is similar to the findings of other studies [1720], suggesting that ANXA1 has compensatory anti-inflammatory effects in asthma. But increased plasma ANXA1 level in stable asthma is decreased in exacerbated asthmatics, indicating that circulating ANXA1 may be decreased due to ANXA1 increase in target inflammatory site in exacerbated state of asthma. To clarify the effect of steroids and particulate matter on ANXA1, we determined the effect of GCs and particulate matter on ANXA1 expression. Dexamethasone and particulate matter induced ANXA1 protein expression, indicating that particulate matter and GCs can activate ANXA1.

Conclusions

In conclusion, this study demonstrated higher levels of ANXA1 in asthmatic patients and lower levels in exacerbated asthmatic patients. In addition, there was a significant relationship between ANXA1 and lung function in asthmatic patients, indicating that ANXA1 is potentially involved in the pathogenesis of asthma.
Furthermore, the findings in this study indicated that serum ANXA1 concentration represents a biomarker for asthma, which has potential utility as a diagnostic tool. However, larger numbers of asthmatic patients are required for prospective studies, and further studies are warranted to investigate the potential mechanism of ANXA1 in asthma.

Acknowledgements

The authors would like to thank the biobank of Soonchunhyang University Bucheon Hospital, a member of the Korea Biobank Network, Republic of Korea, for their assistance in providing the data and biospecimens for this study.

Funding

This research was supported by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant number: HI15C2032) and Soonchunhyang University Research Fund.

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request. The detailed clinical data is not available because of patients’ confidentiality.
All relevant study protocols, the biospecimens and clinical data were approved by the Ethics Committee of Soonchunhyang University Hospital, a member of the Korea Biobank Network (approval no. SCHBC-IRB-2014-10-011). Informed written consent was obtained from each patient prior to inclusion in this study. This study of animals was approved by Soonchunhyang University’s institutional animal care and use committee.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
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.
Literatur
1.
Zurück zum Zitat Fatimathas L, Moss SE. Annexins as disease modifiers. Histol Histopathol. 2010 Apr;25(4):527–32.PubMed Fatimathas L, Moss SE. Annexins as disease modifiers. Histol Histopathol. 2010 Apr;25(4):527–32.PubMed
2.
Zurück zum Zitat Bizzarro V, Fontanella B, Franceschelli S, Pirozzi M, Christian H, Parente L, et al. Role of annexin A1 in mouse myoblast cell differentiation. J Cell Physiol. 2010;224(3):757–65.CrossRefPubMed Bizzarro V, Fontanella B, Franceschelli S, Pirozzi M, Christian H, Parente L, et al. Role of annexin A1 in mouse myoblast cell differentiation. J Cell Physiol. 2010;224(3):757–65.CrossRefPubMed
3.
Zurück zum Zitat Bizzarro V, Petrella A, Parente L. Annexin A1: novel roles in skeletal muscle biology. J Cell Physiol. 2012;227(8):3007–15.CrossRefPubMed Bizzarro V, Petrella A, Parente L. Annexin A1: novel roles in skeletal muscle biology. J Cell Physiol. 2012;227(8):3007–15.CrossRefPubMed
4.
Zurück zum Zitat Bizzarro V, Fontanella B, Carratù A, Belvedere R, Marfella R, Parente L, et al. Annexin A1 N-terminal derived peptide Ac2-26 stimulates fibroblast migration in high glucose conditions. PLoS One. 2012;7(9):e45639.CrossRefPubMedPubMedCentral Bizzarro V, Fontanella B, Carratù A, Belvedere R, Marfella R, Parente L, et al. Annexin A1 N-terminal derived peptide Ac2-26 stimulates fibroblast migration in high glucose conditions. PLoS One. 2012;7(9):e45639.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Bizzarro V, Belvedere R, Dal Piaz F, Parente L, Petrella A. Annexin A1 induces skeletal muscle cell migration acting through formyl peptide receptors. PLoS One. 2012;7(10):e48246.CrossRefPubMedPubMedCentral Bizzarro V, Belvedere R, Dal Piaz F, Parente L, Petrella A. Annexin A1 induces skeletal muscle cell migration acting through formyl peptide receptors. PLoS One. 2012;7(10):e48246.CrossRefPubMedPubMedCentral
6.
7.
8.
Zurück zum Zitat Monastyrskaya K, Babiychuk EB, Draeger A. The annexins: spatial and temporal coordination of signaling events during cellular stress. Cell Mol Life Sci. 2009;66(16):2623–42.CrossRefPubMed Monastyrskaya K, Babiychuk EB, Draeger A. The annexins: spatial and temporal coordination of signaling events during cellular stress. Cell Mol Life Sci. 2009;66(16):2623–42.CrossRefPubMed
9.
Zurück zum Zitat Hayes MJ, Rescher U, Gerke V, Moss SE. Annexin-actin interactions. Traffic. 2004;5(8):571–6.CrossRefPubMed Hayes MJ, Rescher U, Gerke V, Moss SE. Annexin-actin interactions. Traffic. 2004;5(8):571–6.CrossRefPubMed
10.
Zurück zum Zitat Belvedere R, Bizzarro V, Popolo A, Dal Piaz F, Vasaturo M, Picardi P, et al. Role of intracellular and extracellular annexin A1 in migration and invasion of human pancreatic carcinoma cells. BMC Cancer. 2014;14:961.CrossRefPubMedPubMedCentral Belvedere R, Bizzarro V, Popolo A, Dal Piaz F, Vasaturo M, Picardi P, et al. Role of intracellular and extracellular annexin A1 in migration and invasion of human pancreatic carcinoma cells. BMC Cancer. 2014;14:961.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Ye RD, Boulay F, Wang JM, Dahlgren C, Gerard C, Parmentier M, et al. International Union of Basic and Clinical Pharmacology. LXXIII. Nomenclature for the formyl peptide receptor (FPR) family. Pharmacol Rev. 2009;61(2):119–61.CrossRefPubMedPubMedCentral Ye RD, Boulay F, Wang JM, Dahlgren C, Gerard C, Parmentier M, et al. International Union of Basic and Clinical Pharmacology. LXXIII. Nomenclature for the formyl peptide receptor (FPR) family. Pharmacol Rev. 2009;61(2):119–61.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Ferlazzo V, D'Agostino P, Milano S, Caruso R, Feo S, Cillari E, et al. Anti-inflammatory effects of annexin-1: stimulation of IL-10 release and inhibition of nitric oxide synthesis. Int Immunopharmacol. 2003;3(10–11):1363–9.CrossRefPubMed Ferlazzo V, D'Agostino P, Milano S, Caruso R, Feo S, Cillari E, et al. Anti-inflammatory effects of annexin-1: stimulation of IL-10 release and inhibition of nitric oxide synthesis. Int Immunopharmacol. 2003;3(10–11):1363–9.CrossRefPubMed
13.
Zurück zum Zitat Parente L, Solito E. Annexin 1: more than an anti-phospholipase protein. Inflamm Res. 2004;53(4):125–32.CrossRefPubMed Parente L, Solito E. Annexin 1: more than an anti-phospholipase protein. Inflamm Res. 2004;53(4):125–32.CrossRefPubMed
14.
Zurück zum Zitat Smith SF, Tetley TD, Guz A, Flower RJ. Detection of lipocortin 1 in human lung lavage fluid: lipocortin degradation as a possible proteolytic mechanism in the control of inflammatory mediators and inflammation. Environ Health Perspect. 1990;85:135–44.CrossRefPubMedPubMedCentral Smith SF, Tetley TD, Guz A, Flower RJ. Detection of lipocortin 1 in human lung lavage fluid: lipocortin degradation as a possible proteolytic mechanism in the control of inflammatory mediators and inflammation. Environ Health Perspect. 1990;85:135–44.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Vishwanatha JK, Davis RG, Rubinstein I, Floreani A. Annexin I degradation in bronchoalveolar lavage fluids from healthy smokers: a possible mechanism of inflammation. Clin Cancer Res Clin Cancer Res. 1998 Oct;4(10):2559–64.PubMed Vishwanatha JK, Davis RG, Rubinstein I, Floreani A. Annexin I degradation in bronchoalveolar lavage fluids from healthy smokers: a possible mechanism of inflammation. Clin Cancer Res Clin Cancer Res. 1998 Oct;4(10):2559–64.PubMed
16.
Zurück zum Zitat Ng FS, Wong KY, Guan SP, Mustafa FB, Kajiji TS, Bist P, et al. Annexin-1-deficient mice exhibit spontaneous airway hyperresponsiveness and exacerbated allergen-specific antibody responses in a mouse model of asthma. Clin Exp Allergy. 2011;41(12):1793–803.CrossRefPubMed Ng FS, Wong KY, Guan SP, Mustafa FB, Kajiji TS, Bist P, et al. Annexin-1-deficient mice exhibit spontaneous airway hyperresponsiveness and exacerbated allergen-specific antibody responses in a mouse model of asthma. Clin Exp Allergy. 2011;41(12):1793–803.CrossRefPubMed
17.
Zurück zum Zitat Eke Gungor H, Tahan F, Gokahmetoglu S, Saraymen B. Decreased levels of lipoxin A4 and annexin A1 in wheezy infants. Int Arch Allergy Immunol. 2014;163(3):193–7.CrossRefPubMed Eke Gungor H, Tahan F, Gokahmetoglu S, Saraymen B. Decreased levels of lipoxin A4 and annexin A1 in wheezy infants. Int Arch Allergy Immunol. 2014;163(3):193–7.CrossRefPubMed
18.
Zurück zum Zitat Levy BD, Bonnans C, Silverman ES, Palmer LJ, Marigowda G, Israel E. Diminished lipoxin biosynthesis in severe asthma. Am J Respir Crit Care Med. 2005;172(7):824–30.CrossRefPubMedPubMedCentral Levy BD, Bonnans C, Silverman ES, Palmer LJ, Marigowda G, Israel E. Diminished lipoxin biosynthesis in severe asthma. Am J Respir Crit Care Med. 2005;172(7):824–30.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Planagumà A, Kazani S, Marigowda G, Haworth O, Mariani TJ, Israel E, et al. Airway lipoxin A4 generation and lipoxin A4 receptor expression are decreased in severe asthma. Am J Respir Crit Care Med. 2008;178(6):574–82.CrossRefPubMedPubMedCentral Planagumà A, Kazani S, Marigowda G, Haworth O, Mariani TJ, Israel E, et al. Airway lipoxin A4 generation and lipoxin A4 receptor expression are decreased in severe asthma. Am J Respir Crit Care Med. 2008;178(6):574–82.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Vachier I, Bonnans C, Chavis C, Farce M, Godard P, Bousquet J, et al. Severe asthma is associated with a loss of LX4, an endogenous anti-inflammatory compound. J Allergy Clin Immunol. 2005;115(1):55–60.CrossRefPubMed Vachier I, Bonnans C, Chavis C, Farce M, Godard P, Bousquet J, et al. Severe asthma is associated with a loss of LX4, an endogenous anti-inflammatory compound. J Allergy Clin Immunol. 2005;115(1):55–60.CrossRefPubMed
22.
Zurück zum Zitat Moon KY, Lee PH, Kim BG, Park CS, Leikauf GD, Jang AS. Claudin 5 in a murine model of allergic asthma: Its implication and response to steroid treatment. J Allergy Clin Immunol. 2015; 136(6):1694–6.e1–5 Moon KY, Lee PH, Kim BG, Park CS, Leikauf GD, Jang AS. Claudin 5 in a murine model of allergic asthma: Its implication and response to steroid treatment. J Allergy Clin Immunol. 2015; 136(6):1694–6.e1–5
23.
Zurück zum Zitat Bouter A, Carmeille R, Gounou C, Bouvet F, Degrelle SA, Evain-Brion D, et al. Review: Annexin-A5 and cell membrane repair. Placenta. 2015 Apr;36(Suppl 1):S43–9.CrossRefPubMed Bouter A, Carmeille R, Gounou C, Bouvet F, Degrelle SA, Evain-Brion D, et al. Review: Annexin-A5 and cell membrane repair. Placenta. 2015 Apr;36(Suppl 1):S43–9.CrossRefPubMed
24.
Zurück zum Zitat Blackwell GJ, Carnuccio R. Di RosaM, flower RJ, Parente L, Persico P. A polypeptide causing the anti-phospholipase effect of glucocorticoids. Nature. 1980;287(5778):147–9.CrossRefPubMed Blackwell GJ, Carnuccio R. Di RosaM, flower RJ, Parente L, Persico P. A polypeptide causing the anti-phospholipase effect of glucocorticoids. Nature. 1980;287(5778):147–9.CrossRefPubMed
25.
Zurück zum Zitat Miele L, Cordella-Miele E, Facchiano A, Mukherjee AB. Novel anti-inflammatory peptides from the region of highest similarity between uteroglobin and lipocortin I. Nature. 1988;335(6192):726–30.CrossRefPubMed Miele L, Cordella-Miele E, Facchiano A, Mukherjee AB. Novel anti-inflammatory peptides from the region of highest similarity between uteroglobin and lipocortin I. Nature. 1988;335(6192):726–30.CrossRefPubMed
26.
Zurück zum Zitat Jia Y, Morand EF, Song W, Cheng Q, Stewart A, Yang YH. Regulation of lung fibroblast activation by annexin A1. J Cell Physiol. 2013;228(2):476–84.CrossRefPubMed Jia Y, Morand EF, Song W, Cheng Q, Stewart A, Yang YH. Regulation of lung fibroblast activation by annexin A1. J Cell Physiol. 2013;228(2):476–84.CrossRefPubMed
27.
Zurück zum Zitat Perretti M, Christian H, Wheller SK, Aiello I, Mugridge KG, Morris JF, et al. Annexin I is stored within gelatinase granules of human neutrophil and mobilized on the cell surface upon adhesion but not phagocytosis. Cell Biol Int. 2000;24(3):163–74.CrossRefPubMed Perretti M, Christian H, Wheller SK, Aiello I, Mugridge KG, Morris JF, et al. Annexin I is stored within gelatinase granules of human neutrophil and mobilized on the cell surface upon adhesion but not phagocytosis. Cell Biol Int. 2000;24(3):163–74.CrossRefPubMed
28.
Zurück zum Zitat Mulla A, Leroux C, Solito E, Buckingham JC. Correlation between the anti-inflammatory protein annexin 1 (lipocortin 1) and serum cortisol in subjects with normal and dysregulated adrenal function. J Clin Endocrinol Metab. 2005;90(1):557–62.CrossRefPubMed Mulla A, Leroux C, Solito E, Buckingham JC. Correlation between the anti-inflammatory protein annexin 1 (lipocortin 1) and serum cortisol in subjects with normal and dysregulated adrenal function. J Clin Endocrinol Metab. 2005;90(1):557–62.CrossRefPubMed
29.
Zurück zum Zitat Perretti M, Chiang N, La M, Fierro IM, Marullo S. Getting SJ, et al. endogenous lipid- and peptide-derived anti-inflammatory pathways generated with glucocorticoid and aspirin treatment activate the lipoxin A4 receptor. Nat Med. 2002;8(11):1296–302.CrossRefPubMedPubMedCentral Perretti M, Chiang N, La M, Fierro IM, Marullo S. Getting SJ, et al. endogenous lipid- and peptide-derived anti-inflammatory pathways generated with glucocorticoid and aspirin treatment activate the lipoxin A4 receptor. Nat Med. 2002;8(11):1296–302.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Fiore S, Maddox JF, Perez HD, Serhan CN. Identification of a human cDNA encoding a functional high affinity lipoxin A4 receptor. J Exp Med. 1994;180(1):253–60.CrossRefPubMed Fiore S, Maddox JF, Perez HD, Serhan CN. Identification of a human cDNA encoding a functional high affinity lipoxin A4 receptor. J Exp Med. 1994;180(1):253–60.CrossRefPubMed
31.
Zurück zum Zitat Le Y, Gong W, Li B, Dunlop NM, Shen W, Su SB, et al. Utilization of two seven-transmembrane, G protein-coupled receptors, formyl peptide receptor-like 1 and formyl peptide receptor, by the synthetic hexapeptide WKYMVm for human phagocyte activation. J Immunol. 1999;163(12):6777–84.PubMed Le Y, Gong W, Li B, Dunlop NM, Shen W, Su SB, et al. Utilization of two seven-transmembrane, G protein-coupled receptors, formyl peptide receptor-like 1 and formyl peptide receptor, by the synthetic hexapeptide WKYMVm for human phagocyte activation. J Immunol. 1999;163(12):6777–84.PubMed
32.
Zurück zum Zitat VanCompernolle SE, Clark KL, Rummel KA, Todd SC. Expression and function of formyl peptide receptors on human fibroblast cells. J Immunol. 2003;171(4):2050–6.CrossRefPubMed VanCompernolle SE, Clark KL, Rummel KA, Todd SC. Expression and function of formyl peptide receptors on human fibroblast cells. J Immunol. 2003;171(4):2050–6.CrossRefPubMed
33.
Zurück zum Zitat Sawmynaden P, Perretti M. Glucocorticoid upregulation of the annexin-A1 receptor in leukocytes. Biochem Biophys Res Commun. 2006;349(4):1351–5.CrossRefPubMed Sawmynaden P, Perretti M. Glucocorticoid upregulation of the annexin-A1 receptor in leukocytes. Biochem Biophys Res Commun. 2006;349(4):1351–5.CrossRefPubMed
Metadaten
Titel
Annexin A1 in plasma from patients with bronchial asthma: its association with lung function
verfasst von
Sun-Hye Lee
Pureun-Haneul Lee
Byeong-Gon Kim
Hyun-Jeong Seo
Ae-Rin Baek
Jong-Sook Park
June-Hyuk Lee
Sung-Woo Park
Do-Jin Kim
Choon-Sik Park
An-Soo Jang
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
BMC Pulmonary Medicine / Ausgabe 1/2018
Elektronische ISSN: 1471-2466
DOI
https://doi.org/10.1186/s12890-017-0557-5

Weitere Artikel der Ausgabe 1/2018

BMC Pulmonary Medicine 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Triglyzeridsenker schützt nicht nur Hochrisikopatienten

10.05.2024 Hypercholesterinämie Nachrichten

Patienten mit Arteriosklerose-bedingten kardiovaskulären Erkrankungen, die trotz Statineinnahme zu hohe Triglyzeridspiegel haben, profitieren von einer Behandlung mit Icosapent-Ethyl, und zwar unabhängig vom individuellen Risikoprofil.

Gibt es eine Wende bei den bioresorbierbaren Gefäßstützen?

In den USA ist erstmals eine bioresorbierbare Gefäßstütze – auch Scaffold genannt – zur Rekanalisation infrapoplitealer Arterien bei schwerer PAVK zugelassen worden. Das markiert einen Wendepunkt in der Geschichte dieser speziellen Gefäßstützen.

Vorsicht, erhöhte Blutungsgefahr nach PCI!

10.05.2024 Koronare Herzerkrankung Nachrichten

Nach PCI besteht ein erhöhtes Blutungsrisiko, wenn die Behandelten eine verminderte linksventrikuläre Ejektionsfraktion aufweisen. Das Risiko ist umso höher, je stärker die Pumpfunktion eingeschränkt ist.

Wie managen Sie die schmerzhafte diabetische Polyneuropathie?

10.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Mit Capsaicin-Pflastern steht eine neue innovative Therapie bei schmerzhafter diabetischer Polyneuropathie zur Verfügung. Bei therapierefraktären Schmerzen stellt die Hochfrequenz-Rückenmarkstimulation eine adäquate Option dar.

Update Innere Medizin

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