Skip to main content
Erschienen in: BMC Pulmonary Medicine 1/2017

Open Access 01.12.2017 | Research article

Identification of the lipid biomarkers from plasma in idiopathic pulmonary fibrosis by Lipidomics

verfasst von: Feng Yan, Zhensong Wen, Rui Wang, Wenling Luo, Yufeng Du, Wenjun Wang, Xianyang Chen

Erschienen in: BMC Pulmonary Medicine | Ausgabe 1/2017

Abstract

Background

Idiopathic pulmonary fibrosis (IPF) is an irreversible interstitial pulmonary disease featured by high mortality, chronic and progressive course, and poor prognosis with unclear etiology. Currently, more studies have been focusing on identifying biomarkers to predict the progression of IPF, such as genes, proteins, and lipids. Lipids comprise diverse classes of molecules and play a critical role in cellular energy storage, structure, and signaling. The role of lipids in respiratory diseases, including cystic fibrosis, asthma and chronic obstructive pulmonary disease (COPD) has been investigated intensely in the recent years. The human serum lipid profiles in IPF patients however, have not been thoroughly understood and it will be very helpful if there are available molecular biomarkers, which can be used to monitor the disease progression or provide prognostic information for IPF disease.

Methods

In this study, we performed the ultraperformance liquid chromatography coupled with quadrupole time of flight mass spectrometry (UPLC-QTOF/MS) to detect the lipid variation and identify biomarker in plasma of IPF patients. The plasma were from 22 IPF patients before received treatment and 18 controls.

Results

A total of 507 individual blood lipid species were determined with lipidomics from the 40 plasma samples including 20 types of fatty acid, 159 types of glycerolipids, 221 types of glycerophospholipids, 47 types of sphingolipids, 46 types of sterol lipids, 7 types of prenol lipids, 3 types of saccharolipids, and 4 types of polyketides. By comparing the variations in the lipid metabolite levels in IPF patients, a total of 62 unique lipids were identified by statistical analysis including 24 kinds of glycerophoslipids, 30 kinds of glycerolipids, 3 kinds of sterol lipids, 4 kinds of sphingolipids and 1 kind of fatty acids. Finally, 6 out of 62 discriminating lipids were selected as the potential biomarkers, which are able to differentiate between IPF disease and controls with ROC analysis.

Conclusions

Our results provided vital information regarding lipid metabolism in IPF patients and more importantly, a few potentially promising biomarkers were firstly identified which may have a predictive role in monitoring and diagnosing IPF disease.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12890-017-0513-4) contains supplementary material, which is available to authorized users.
Zhensong Wen is also a first co-author
Abkürzungen
CLs
Cardiolipin
IPF
Idiopathic pulmonary fibrosis
OPLS-DA
Orthogonal partial least squares discriminant analysis
PAs
Glycerophosphatidic acids
PCs
Glycerophosphocholines
PEs
Glycerophosphoethanolamines
PGs
Glycerophosphoglycerols
PIs
Glycerophosphoinositols
PSs
Glycerophosphoserines
ROC
Receiver operating characteristic
UPLC-QTOF/MS
Ultraperformance liquid chromatography coupled with quadrupole time of flight mass spectrometry
VIP
Variable importance for projection

Background

Idiopathic pulmonary fibrosis (IPF) is a disease featured as chronic, progressive, irreversible interstitial pneumonia with a poor prognosis of unknown etiology [1] and a median survival of 3 to 5 years after initial diagnosis [2, 3]. It more commonly occurs in patients of 50 to 70 years of age. This disease is characterized by the histological pattern of usual interstitial pneumonia [4, 5] with parenchymal fibrosis and excess collagen deposition [6]. It is well accepted that the pathology of this disease includes fibroblast/myofibroblast proliferation, activation of alveolar epithelial cells, with exacerbated deposit of extracellular matrix leading to the gradual destruction of the lung tissue [7]. Although there have been increasing number of studies investigating the pathogenesis of idiopathic pulmonary fibrosis (IPF), the lacking of effective treatments and early diagnostic tools indicate the urgent needs for reliable biomarkers in both diagnosing at early stage and monitoring the progression of this disease. In the recent publications, a few potentially useful blood cellular and molecular biomarkers have been identified, including chemokines, proteases and growth factors [8]. Despite the recent progress, there has not been a single biomarker proved to be useful in diagnosing and monitoring the progress of IPF. The diagnosis of IPF has not changed much over the last few years, which means that a multi-disciplinary approach is probably required for a breakthrough. With the new emergent technology of lipidomics, to identify novel lipid biomarkers for diagnostic and monitor purposes of IPF becomes possible.
Lipids comprise diverse classes of molecules that play a critical role in cellular energy storage, structure, and signaling [911]. Previously, lipids are only considered as the components of membranes and source. Now lipids are known to act as a indispensable factor in the immune response by organizing signaling complexes in cellular membrane, such as lipid rafts [12] or affecting the immune reaction by release of lipid-derived mediators [13, 14]. The etiologies of certain diseases have been proven to be associated with individual lipid molecules and many studies have indicated that certain lipid metabolic disorders or abnormalities can lead to a variety of human diseases [10, 1519].
The role of lipids in lung and respiratory diseases has attracted more attention in recent years including cystic fibrosis, asthma and COPD which are all associated with abnormal metabolism. For example, the epithelium lipid metabolism has been proved to be changed in asthmatic patients [20, 21]. The amount of ceramides in the airway epithelium of a guinea pig model was found to be increased in response to the induction of experimental allergic asthma [22]. In another study, the increased amount of ceramides was detected in the airway epithelium and has been linked to cell death, infection susceptibility and immune inflammation in cystic fibrosis [23]. When it comes to IPF however, the human plasma lipid profiles of IPF is so far poorly understood and to identify reliable and unique lipid molecular biomarkers will be very beneficial in IPF diagnosis and management [24]. Based on the above reasons, to analyze and identify potential IPF-specific lipid biomarkers will contribute significantly to the diagnosis and management in IPF patients.
With the development of omicis [25] and the advanced mass spectrometry have made it feasible to identify and quantify a variety of lipids species in human samples such as the tandem mass spectrometer utilized (MS/MS) [26, 27],direct infusion MS (DIMS) [28], and liquid chromatography-mass spectrometry (LC-MS) [2932]. Among different LC-MS platforms, ultraperformance liquid chromatography coupled with quadrupole time of flight mass spectrometry (UPLC-QTOF/MS) is widely adapted to lipidomics due to its enhanced reproducibility of retention time [3335]. In this study, a global lipid profiling was performed containing measurement of 20 kinds of fatty acid, 159 kinds of glycerolipids, 221 kinds of glycerophospholipids, 47 kinds of sphingolipids, 46 kinds of sterol lipids, 7 kinds of prenol lipids, 3 kinds of saccharolipids, and 4 kinds of polyketides. Subsequently, the correlation analysis, receiver operating characteristic (ROC) analysis and orthogonal partial least squares discriminant analysis (OPLS-DA) were performed to evaluate the variations in lipid metabolites. The potential influence of gender, smoking history, and disease stages on the lipid metabolites was also looked at between IPF object and/or controls. Our results provided vital information regarding lipid metabolism in IPF patients and more importantly, a few potentially promising biomarkers were firstly identified which may have a predictive role in monitoring and diagnosing IPF disease.

Methods

Sample and collection

In this study, 22 IPF patients and 18 controls were obtained from the First Hospital of Tsinghua University from January 2014 to March 2016. All IPF patients were diagnosed with IPF after the age of 60, and all of controls were all above 60 years old when examined. The demographic figures are listed in the Table 1. The stages of disease are classified into mild and severe according to the arterial blood oxygen partial pressure (≧ 60 mmHg). The diagnosis of IPF patients was made according to the internal recommendations of the ATS/ERS/JRS/ALAT statement using high-resolution computed tomography (HRCT), as well as the clinical history of the patient [36]. All cases were discussed in our discussion team about interstitial lung disease composed of: a specialist in pulmonary rehabilitation, a specialist in occupational medicine, a radiologist, a rheumatologist, a pulmonologist and a pathologist. Blood samples were collected from each patient before received treatment and control. The controls were precluded if there was a history of pulmonary disease. For each subject, 10 ml whole blood was collected into a vessel tube containing heparin as anticoagulant. Each sample was centrifuged at 1500 x g for 15 min to collect serum and stored at -80 °C immediately until further analysis.
Table 1
Sample demographics of the study subjects
 
IPF (N = 22)
Control (N = 18)
Age, mean
73
72
Gender (males/females)
11/11
8/10
Smoking History (Yes/No)
9/13
10/8
Stages (mild/severe)
11/11

Lipidomics

Liquid chromatography-mass spectrometry (LC-MS)-grade isopropanol, acetonitrile, methanol and water were purchased from Fisher Scientific (New Jersey, USA). Debrisoquine, Pro-Asn, glycoursodeoxycholic acid andmalic acid, 4-nitrobenzoic acid (4-NBA) were products of Sigma (St. Louis, MO, USA). High purity formic acid (99%) was provided by Thermo-Scientific (Rockford, IL). The serum lipid extraction was conducted as described previously [37]. Briefly, the plasma samples were thawed on ice before being vortexed. For metabolite extraction, 25 μL of plasma sample was mixed with 175 μL of extraction buffer (25% acetonitrile in 40% methanol and 35% water). The sample was then incubated on ice for 10 min before centrifuged again at 14,000 rpm at 4 °C for 20 min. Subsequently, the produced supernatant was transferred to a fresh tube and dried under vacuum. After drying, the dried samples were reconstituted in 200 μL of buffer containing 5% methanol, 1% acetonitrile and 94% water. Fine particles were removed by centrifuge at 13,000 rpm for 20 min at 4 °C. Finally, the supernatant was transferred to a glass vial for ultraperformance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-QTOF-MS) analysis.

Statistical analysis

All statistical analyses were conducted using R software version 2.9.1. The OPLS score plots and T-test and variable importance for projection (VIP) statistics were used to select significant variables leading to group separation. A supervised OPLS analysis was applied in our study to identify potential lipids that were used to classify the samples and remove non-correlated variables. The differences between the intensities of lipids in IPF and healthy controls were compared by T test when the data follow a normal distribution or Wilcoxon rank-sum test when otherwise. In this study, the identified lipids were pre-selected as potential biomarkers when VIP value is bigger than 1.0. To analyze the diagnostic value of potential lipid biomarkers for identifying IPF disease, a ROC analysis was performed. Correlation analysis of differential lipids was performed by MetaboAnalyst software. The influence of gender, smoking history, and stages of disease of individual subjects on lipid metabolism were evaluated by the Mann-Whitney U-test. The p values of less than 0.05 was considered statistically significant.

Ethics statement

The clinical IPF samples included in this study were collected from the First hospital of Tsinghua University. All patient data were anonymous, so informed consent for participation was not required. The use of these samples was approved by the Institutional Review Board for human studies at the First Hospital of Tsinghua University. In this study, all personal information including name, date of birth, and contact information was all de-identified and not disclosed.

Results

IPF plasma lipid profiles of 507 apparent lipid species

In this study, the plasma lipid profiles of 507 individual lipid species were determined with lipidomics from 40 plasma samples; 22 from IPF patients with 18 controls (Additional file 1: Table S1). The detected individual plasma apparent lipid species were classified into 8 categories: fatty acid, glycerolipid, glycerophospholipid, sphingolipid, sterol lipid, prenol lipid, saccharolipid, and polyketide in accordance with NIH-funded Consortium, which has built an ongoing website tools offering precise information based on numerous lipidomics studies [38]. The 507 individual lipid species included 20 kinds of fatty acyls (3.94%), 159 kinds of glycerolipids (31.36%), 221 kinds of glycerophospholipids (43.59%), 47 kinds of sphingolipids (9.27%), 46 kinds of sterol lipids (9.07%), 7 kinds of prenol lipids (1.38%), 3 kinds of saccharolipids (0.59%), and 4 kinds of polyketides (0.79%), respectively. The significant differences of each individual apparent lipid species between IPF patients and control groups were detailed in the context of this study.

Statistical analysis of the lipid profiling

Supervised orthogonal partial least squares (OPLS) analysis identifed the biggest variation in lipid profiling using a few orthogonal latent variables and was performed with the lipid-obtained data on the plasma in positive ion mode by the UPLC-QTOF-MS/MS. The metabolic patterns were plotted by the OPLS-DA model (Fig. 1A). The OPLS-DA model was used to unfold the difference of plasma metabolic pattern between IPF patients and control group. The OPSL score plot revealed the significant deviation between IPF patients and controls.
In order to identify the potential biomarkers, the S-plot analysis was used based on the plasma lipid profiling data (Fig. 1B). By comparing changes in the lipid metabolite levels of IPF patients, a total of 62 unique lipids were detected [variable importance for projection (VIP > 1), Table 2]. The lipids were identified according to the reported methods as described previously [37]. They included 24 kinds of glycerophoslipids, 30 kinds of glycerolipids, 3 kinds of sterol lipids, 4 kinds of sphingolipids and 1 kind of fatty acid. The above lipids were selected as potential biomarkers from the S-plot for further analysis in our study.
Table 2
List of assigned statistically significantly lipid molecules and change trend of biomarkers on plasma lipid profile in positive ion mode after comparison of IPF patient samples to control samples
No.
Code
Calc m/z
tR
VIP
Trenda
Accepted Description
LipidIon
Element
Lipid class
P-Value
1
R1
1516.124
5.06
8.58719
CL(1′-[20:0/20:0],3′-[18:1/18:2])
M + H,M + Na,M + H + Na
C85H160O17P2
Glycerophospholipids
7.90E-08***
2
R2
872.769
14.54
7.63644
TG(16:0/18:2/18:2)[iso3]
M + H,M + NH4
C55H98O6
Glycerolipids
1.24E-07***
3
R3
806.567
4.52
6.33481
PC(22:6/16:0)
M + H,M + Na,M + K,2 M + H,2 M + Na
C46H80NO8P
Glycerophospholipids
9.24E-07***
4
877.724
14.55
6.05829
TG(16:1/18:1/18:2)[iso6]
M + Na,M + K,2 M + Na
C55H98O6
Glycerolipids
0.2370
5
874.785
14.82
4.60354
TG(16:0/18:1/18:2)[iso6]
M + H-H2O,M + H,M + NH4,M + Na,
M + K,2 M + NH4,2 M + Na
C55H100O6
Glycerolipids
0.1117
6
R4
782.567
4.23
4.39906
PA(17:2/22:1)
M + ACN + H
C42H77O8P
Glycerophospholipids
0.0074**
7
903.739
14.55
3.8738
TG(16:1/18:1/20:3)[iso6]
M + Na,M + K
C57H100O6
Glycerolipids
0.0017**
8
R5
900.801
14.84
3.45942
TG(16:0/18:1/20:3)[iso6]
M + NH4,M + Na
C57H102O6
Glycerolipids
0.0174*
9
R6
902.816
15.11
3.44338
TG(16:0/18:3/20:0)[iso6]
M + NH4,M + Na,2 M + Na
C57H104O6
Glycerolipids
0.0035**
10
R7
666.618
14.64
3.39521
Stigmasteryl ester(16:1)
M + H,M + NH4,M + K,2 M + NH4,2 M + Na
C45H76O2
Stero lipids
7.79E-08***
11
R8
834.598
5.99
3.14588
PS(20:0/19:0)
M + H-2H2O,M + H,M + Na,2 M + H,2 M + Na
C45H88NO10P
Glycerophospholipids
0.0019**
12
R9
369.352
14.65
3.00377
(E,E)-3,7,11-Trimethyl-2,
6,10-dodecatrienyl dodecanoate
M + H-2H2O
C27H48O2
Fatty acids
7.31E-08***
13
R10
577.519
15.08
2.65073
DG(16:0/18:1/0:0)
M + H-H2O
C37H70O5
Glycerolipids
7.02E-08***
14
R11
848.769
14.77
2.49731
TG(16:0/16:1/18:1)[iso6]
M + H,M + NH4,M + Na,M + K
C53H98O6
Glycerolipids
0.0174*
15
R12
780.552
4.07
2.47828
PE(20:0/17:2)
M + Na
C42H80NO8P
Glycerophospholipids
0.0232*
16
R13
603.535
15.07
2.44092
DG(O-16:0/18:1)
M + Na
C37H72O4
Glycerolipids
7.02E-08***
17
758.569
3.96
2.34541
PC(16:0/18:2)
M + H
C42H80NO8P
Glycerophospholipids
0.5407
18
R14
701.558
3.58
2.32011
SM(d16:1/18:1)
M + H,M + Na,M + K,2 M + H,2 M + Na
C39H77N2O6P
Sphingolipids
0.0006***
19
R15
496.339
0.75
2.29702
PC(0:0/16:0)
M + H-H2O,M + H,M + Na,M + K,2 M + K
C24H50NO7P
Glycerophospholipids
0.0049**
20
R16
369.352
15.00
2.27907
3-Deoxyvitamin D3
M + H,M + NH4,2 M + H
C27H44
Stero lipids
7.31E-08***
21
870.753
14.27
2.16619
TG(14:1/16:0/22:4)[iso6]
M + NH4,M + Na,M + K
C55H96O6
Glycerolipids
0.2829
22
R17
925.796
14.55
2.1151
TG(18:4/20:3/22:0)[iso6]
M + H-2H2O
C63H108O6
Glycerolipids
7.86E-08***
23
R18
801.682
10.78
2.06029
SM(d17:1/24:0)
M + H,M + Na
C46H93N2O6P
Sphingolipids
1.85E-05***
24
899.709
13.99
1.99376
TG(16:1/18:2/20:4)[iso6]
M + NH4,M + Na,M + K
C57H96O6
Glycerolipids
0.1458
25
R19
546.352
1.24
1.88084
PS(22:0/0:0)
M + H-2H2O
C28H56NO9P
Glycerophospholipids
0.0249*
26
873.693
13.97
1.87046
TG(15:1/17:1/20:4)[iso6]
M + Na,M + K
C55H94O6
Glycerolipids
0.1245
27
834.598
5.14
1.83438
PC(22:4/18:2)
M + H,M + Na,2 M + Na
C48H84NO8P
Glycerophospholipids
0.4226
28
R20
832.580
5.49
1.81376
PC(20:2/18:2)
M + Na,2 M + Na
C46H84NO8P
Glycerophospholipids
1.22E-06***
29
931.771
15.01
1.81008
TG(17:0/17:0/22:5)[iso3]
M + Na
C59H104O6
Glycerolipids
0.5582
30
786.601
4.84
1.76508
PC(18:1/18:1)
M + H-H2O,M + H,M + K
C44H84NO8P
Glycerophospholipids
0.9026
31
1564.121
2.72
1.68796
PC(22:4/14:0)
M + NH4,M + K,2 M + H
C44H80NO8P
Glycerophospholipids
0.2314
32
R21
603.532
14.82
1.64593
DG(18:0/18:2/0:0)[iso2]
M + H-H2O
C39H72O5
Glycerolipids
7.53E-08***
33
R22
575.503
14.54
1.62251
DG(16:0/18:2/0:0)[iso2]
M + H-H2O
C37H68O5
Glycerolipids
2.58E-07***
34
812.614
6.86
1.61868
PS(O-20:0/20:2)
M + H-H2O,M + H
C46H88NO9P
Glycerophospholipids
0.0893
35
849.693
14.21
1.61511
TG(14:1/18:1/18:2)[iso6]
M + NH4,M + Na,M + K
C53H94O6
Glycerolipids
0.2829
36
904.831
15.36
1.53098
TG(17:0/18:1/19:1)[iso6]
M + NH4,M + Na,M + K,2 M + Na
C57H106O6
Glycerolipids
0.1615
37
R23
828.551
5.01
1.5305
PS(P-18:0/19:0)
M + H-H2O,M + K
C43H84NO9P
Glycerophospholipids
0.0428*
38
R24
599.504
14.54
1.49112
DG(18:1/18:3/0:0)[iso2]
M + H-H2O,M + H
C39H68O5
Glycerolipids
1.21E-07***
39
901.725
13.34
1.48742
TG(16:0/16:0/22:6)[iso3]
M + NH4,M + Na
C57H98O6
Glycerolipids
0.7340
40
R25
955.759
13.01
1.48372
TG(19:1/22:6/22:6)[iso3]
M + H-2H2O
C66H102O6
Glycerolipids
3.56E-06***
41
925.724
14.16
1.46671
TG(12:0/22:2/22:6)[iso6]
M + NH4,M + Na,M + K
C59H98O6
Glycerolipids
0.1458
42
879.739
13.99
1.38113
GalCer(d18:1/26:1(17Z))
M + ACN + H
C50H95NO8
Sphingolipids
0.7340
43
R26
601.518
14.53
1.36641
DG(18:1/18:2/0:0)[iso2]
M + H-H2O
C39H70O5
Glycerolipids
8.04E-07***
44
810.599
4.57
1.32667
PC(18:0/18:1)
M + Na
C44H86NO8P
Glycerophospholipids
0.1534
45
R27
759.637
6.96
1.30466
SM(d18:1/20:0)
M + H,M + Na,2 M + H
C43H87N2O6P
Sphingolipids
0.0139*
46
524.371
1.10
1.29548
PC(18:0/0:0)
M + H,M + K
C26H54NO7P
Glycerophospholipids
0.2479
47
R28
830.566
3.80
1.25023
PC(22:6/18:2)
M + H,M + Na
C48H80NO8P
Glycerophospholipids
0.0038**
48
R29
1620.182
4.50
1.22041
PI-Cer(d20:0/16:0)
M + H-2H2O,M + K,2 M + H
C42H84NO11P
Glycerophospholipids
7.90E-08***
49
828.551
4.04
1.20343
PE(22:6/19:0)
M + Na
C46H80NO8P
Glycerophospholipids
0.2060
50
R30
948.800
14.59
1.20004
TG(18:1/20:3/20:4)[iso6]
M + NH4
C61H102O6
Glycerolipids
7.90E-08***
51
R31
794.602
4.86
1.19345
PC(20:3/P-18:1)
M + H,M + Na,2 M + H
C46H84NO7P
Glycerophospholipids
1.93E-07***
52
R32
790.569
5.79
1.18604
PA(20:5/20:1)
M + H-H2O,M + ACN + H
C43H73O8P
Glycerophospholipids
0.0012**
53
881.754
14.25
1.18421
TG(15:0/18:1/19:1)[iso6]
M + Na
C55H102O6
Glycerolipids
0.2829
54
1494.138
6.25
1.17806
CL(1′-[20:0/18:0],3′-[18:0/18:0])
M + H
C83H162O17P2
Glycerophospholipids
0.5751
55
R33
851.708
13.63
1.14176
20:1-Glc-Sitosterol
M + H-H2O
C55H96O7
Stero lipids
0.0161*
56
R34
897.696
13.73
1.10216
TG(18:3/18:3/20:5)[iso3]
M + H
C59H92O6
Glycerolipids
0.0014**
57
542.321
0.60
1.09805
PC(20:5/0:0)
M + H
C28H48NO7P
Glycerophospholipids
0.3318
58
1576.219
8.50
1.05263
PC(17:0/19:1)
M + K,2 M + H
C44H86NO8P
Glycerophospholipids
0.2471
59
951.740
14.31
1.04393
TG(18:4/20:0/20:5)[iso6]
M + Na,M + K
C61H100O6
Glycerolipids
0.3918
60
896.768
14.27
1.01614
TG(18:1/18:2/18:3)[iso6]
M + NH4,M + Na
C57H98O6
Glycerolipids
0.9892
61
R35
920.768
14.30
1.01291
TG(16:0/20:3/20:5)[iso6]
M + NH4,M + K
C59H98O6
Glycerolipids
1.40E-06***
62
905.754
13.68
1.00186
TG(12:0/20:1/22:3)[iso6]
M + Na
C57H102O6
Glycerolipids
0.8811
aChange trend of lipids on IPF patients vs controls. The levels of potential lipid biomarkers labeled with (↑) and (↓) represent up-regulation and down-regulation, respectively. The variable importance for projection (VIP) statistics and T-test were used applied to select significant variables leading to group separation. VIP value larger than 1.0 were considered statistically significant and significant differences in statistics were also defined by P values of < 0.05 (*), < 0.01 (**), and < 0.001 (***)
All the determined glycerophospholipids and sphingolipids showed a decreasing tendency in IPF objects (Table 2). Although all of glycerophospholipids and sphingolipids also showed a similar decreasing pattern in IPF patients, the magnitude of their drop was not the same. 15 out of 30 glycerolipids and 1 out of sterol lipids were lower in IPF objects. On the other hand, the remaining glycerolipids, sterol lipids and fatty acids had an increased level in IPF objects. These findings suggested that the observed changes in lipid profiles were likely caused by different expression in IPF patients.

Correlation and receiver operating characteristic (ROC) curve analysis

To better understand the relationship of metabolite differences with IPF disease, correlation analysis was applied to analyze these identified lipids data (Fig. 2). Firstly, The differences between the intensities of 62 identified lipids in IPF and healthy controls were compared by T test when the data follow a normal distribution or Wilcoxon rank-sum test when otherwise. Comprehensive analysis with P-value, 35 out of 62 identified lipids were selected to further analyze (Table 2, R1 to R35). Then, we evaluated the correlation between IPF disease and 35-selected lipids. As shown in Fig.2, the close correlations can be detected between IPF disease and 12 out of 35 identified lipids, including TG(16:0/18:2/18:2)[iso3] (R2), PC(22:6/16:0) (R3), Stigmasteryl ester(16:1) (R7) and (E,E)-3,7,11-Trimethyl-2,6,10-dodecatrienyl dodecanoate (R9), DG(O-16:0/18:1) (R13), 3-Deoxyvitamin D3 (R16), TG(18:4/20:3/22:0)[iso6] (R17), DG(18:0/18:2/0:0)[iso2] (R21), DG(16:0/18:2/0:0)[iso2] (R22), DG(18:1/18:3/0:0)[iso2] (R24), TG(19:1/22:6/22:6)[iso3] (R25), and DG(18:1/18:2/0:0)[iso2] (R26) (correlation absolute value > 0.7). This result indicates that these individual lipid molecules potentially are useful to differentiated the IPF patients from control group and the accuracy and efficiency possibly can be further increased if more lipid candidates are to be used.
Theoretically, those lipid molecules possessing close correlation with IPF disease would be more promising to be used as biomarkers. To further demonstrate the ability of the 12 out of 35 identified lipids to identify the IPF objects and controls, the receiver operating characteristic (ROC) curve was applied according to the results for the area under the curve (AUC) and sensitivity/specificity at the best cut-off points (Fig. 3). The AUC values of these molecules showed significant differences in the discovery set. 6 (R7, R9, R13, R16, R17 and R21) of them showed higher sensitivity/specificity for identifying IPF objects from controls (Fig. 3). This result have demonstrated that each of the sixmolecules may be used as potential biomarkers in diagnosing IPF disease in the future.

Gender, smoking, and disease status-associated differences in the lipid levels of six-promising biomarkers

To demonstrate whether gender, smoking history, and disease stage have influence on selecting useful future biomarkers, 6 lipid levels in serums from IPF objects and control were further studied. Table 3 summarises the significance of all 6 biomarkers with regards to gender, smoking history, and disease stage-associated differences. None of these 6 was found to have significant differences related to these parameters mentioned above.
Table 3
Differences in the lipid levels of eight-promising biomarkers under the influence of gender, smoking, and disease status. The statistical significance was determined by Mann-Whitney U-test
No.
Gender/P-value
Smoking/P-value
Disease status/P-value
R7
0.92
0.92
0.65
R9
0.51
0.51
0.91
R13
0.81
0.16
0.17
R16
0.40
0.40
0.84
R17
0.85
0.43
0.30
R21
0.96
0.17
0.15

Discussion

IPF is a kind of chronic and progressive disease with low survival rate, and remains to be a clinical challenge. It is regarded as a fetal disease due to its poor prognosis and a low median survival of only 3~5 years after diagnosis. At the moment, only clinical data are available for diagnosis to researchers and clinicians, which is of limited value as they do not reflect the precise pathological mechanisms underlying IPF. To better elucidate disease mechanism and make early diagnosis of IPF, identification of molecular biomarkers with high diagnostic value is paramount. In recent publications, a number of biomarkers have been hypothesized to be present in serum used as potential prognostic or diagnostic tools. The main protein biomarkers associated with cell dysfunction are the Krebs con den lungen-6 (KL-6) antigen and the surfactant protein A and protein D (SP-A and SP-D) [39]. Biomarkers found in IPF involved in fibrogenesis are matrix metalloproteinases-1 and -7 (MMP-1 and MMP-7), which play a role in the breakdown and remodeling of extracellular matrix components [40]. However, no lipid biomarkers have yet been studied for IPF. This work is the first comprehensive investigation into the potential lipid biomarkers for IPF using UPSL-QTOF-MS/MS.
Lipids is the fundamental component of cellular membranes, also exert several essential and critical roles in cellular functions including energy storage, signal transduction, formation of membrane bilayer and cellular barriers. The metabolism of lipid is indicated in numerous human diseases, such as Alzheimer’s disease, diabetes, obesity, atherosclerosis and several types of respiratory diseases. There are eight major categories of lipid types based on their structures: fatty acid, glycerolipid, saccharolipid, polyketide, sphingolipid, sterol lipids, prenol lipid and glycerophospholipid. Although abnormal lipid metabolism has been shown to result in cystic fibrosis and lung injury published by Ollero’s and Goss’ research groups, respectively [41, 42], its potential role in pathology of IPF remains unclear.
In this study, analysis of lipid profile from 22 IPF patients and 18 control subjects revealed the characterization of lipid composition. The glycerophospholipids (GPs) appears to be the important biological molecules for the backbone of cellular membranes. Besides an integral component of biomembranes, GPs also seem to be a reservoir of a large amount of many bioactive mediators [43], which are produced by the reaction of phospholipases on GPs. GPs can be further divided into different categories including glycerophosphoglycerols (PGs), glycerophosphatidic acids (PAs), glycerophosphoinositols (PIs), glycerophosphoserines (PSs), glycerophosphoethanolamines (PEs) and cardiolipin (CLs).
In our study, 2 CLs, 2 PAs, 13 PCs, 2 PEs, 1 PIs, and 4 PSs were identified as unique lipids of IPF patients based on VIP scores (Table 2). The levels of all screened GPs was decreased in IPF patients compared to the control subjects. This phenomenon may be due to the fact that PG is a precursor of CL biosynthesis and PA is the critic substrate for biosynthesis of PI, PG, PE, and PC. A previous TLC-MALDI/TOF-based metabonomics study displayed significantly decreased plasma PCs in cystic fibrosis patients, including PC(P-40:1), PC(38:6), PC(38:5), PC(38:4), PC(38:2), PC(38:0) and PC(36:5) [44]. However, using electrospray ionization tandem mass spectrometry (ESI-MS/MS)-based metabonomics, Marien et al. showed an increase in non-small cell lung cancer of PI(38:3), PI(40:3), PI(38:2), PS(32:0), PS(36:4), PS(36:1), PS(40:2), PS(38:1), PS(34:0), PS(40:1), PS(38:5), PS(34:2) and PS(38:4), compared to normal lung tissues [45]. Increasing evidence implicates that GPs biomarkers were several in agreement with abnormal GPs metabolism found in above mentioned lung disease and IPF model. This result indicated that GPs with top scores could be used as potential biomarkers for IPF.
Our study identified 159 glycerolipids in IPF patients and 30 out of these 159 have the capacity to differentiate IPF patients from control subjects (VIP > 1, Table 2). Glycerolipids possess long chain fatty acids in ester linkage to the glycerol backbone and were classified into two major groups: diacylglycerol (DG) and triacylglycerol (TG), which are the most abundant lipids found in circulating plasma [46]. For example, all six DGs out of 30 glycerolipids showed similar tendencies to increase in the plasma from IPF patients with variable magnitudes. Interestingly, 9 out of 24 TGs displayed increased levels in IPF patients compared to control subjects. A previous study demonstrated that glycerolipids were significantly reduced in lung tissues of mice, which lacked the p53 oncogene [47]. This result indicates that the abnormal function of glycerolipid was associated with lung disease. Further investigation of the exact roles of glycerolipids in IPF patients would be beneficial.
We detected 47 sphingolipids (SLs) by UPSL-QTOF-MS/MS in plasma samples and found that 4 out of them can distinguish the IPF patients from control subjects. 4 types of SLs (GalCerd18:1/26:1, d18:1/20:0, d16:1/18:1, d17:1/24:0) significantly decreased in plasma of patients with IPF. SLs are ubiquitous cellular membrane components that are implicated in multiple cellular processes including autophagy, apoptosis, differentiation and cell division [48]. Ceramide is generated from either sohingomyelin or de novo sphingolipids synthesis [49] and its upregulation has been found in chronic-obstructive pulmonary disease [50]. In contrast, our results showed decreased SLs serum level in IPF patient, which could be due to the differences between different respiratory diseases. In the future, we will compare SLs levels in serum and bronchoalveolar lavage fluids between respiratory diseases, such as COPD and pneumonia. Although it remains to be equivocal whether differences exist in SLs levels between IPF and other respiratory diseases, our data have provided a direction for future investigation into implications of these SLs in IPF patients.
Moreover, this study identified 46 kinds of serol lipids and 20 kinds of fatty acids. 3 kinds of 46 serol lipids and 1 kind of 20 fatty acids were considered as potential biomarkers. Fatty acids are the most important class of lipids and function as precursors of various bioactive lipid molecules. In our study, fatty acid (E,E)-3,7,11-Trimethyl-2,6,10-dodecatrienyl dodecanoate was shown to be positively correlated with IPF and may distinguish IPF patients from control subjects. Three (3-Deoxyvitamin D3,16:1 Stigmasteryl ester, and 20:1-Glc-Sitosterol) out of 46 sterol lipids identified have been shown to correlate with the IPF. Other studies have shown that plasma cholesterol level was significantly increased in diet-induced hyperlipidomic rats [51], and cholesteryl ester apparent lipid molecular species have high sensitivity, specificity and accuracy in the diagnosis of prostate cancer [10]. Further validation of the specificity of these lipid molecules in IPF patients among respiratory disease would be desirable.
Furthermore, we investigated lipid molecules of 12 metabolites possessing close correlations with IPF disease. 6 of them showed higher sensitivity and specificity for identifying IPF patients from control subjects ROC analysis (Fig. 3). Previous study has demonstrated that age, gender, and smoking status can affect plasma lipid metabolite levels in healthy adults [52, 53]. The impact of gender and smoking on 6 promising biomarker levels were determined in this study and no correlation was found (Table 3). Further validations on whether the 6 identified promising biomarkers has the increased ability to discriminate IPF objects from healthy controls or other respiratory disease are highly suggested.
For the analysis of IPF patients and biomarkers, the potential defect in this study was the small sample size. Another limitation is the lack of a longitudinal study, which made it impossible to observe the clinical impact of the present discovery as no treatment responses can be assessed. Although we have identified promising lipid biomarkers in this study, further validations are necessary to evaluate the specificity of the identified biomarkers for IPF diagnosis. Then, further longitudinal multicenter studies would contribute more to evaluate the real value of lipid biomarkers as diagnostic and prognostic tools. In addition, the lipid biomarkers identified in this study should be compared with known diagnostic and prognostic biomarkers in the future, such as KL-6, SP-A, SP-D. This will provide an insight into a better understanding of the diagnostic and prognostic utility of identified lipid biomarkers. Lastly, more specific lipidomics analyses of these lipid changes in IPF will probably help to better understand the IPF pathology and may contribute to future development of novel therapeutic targets.

Conclusions

In conclusions, our study has yielded important information regarding lipid metabolism in IPF patients and presents the first identification of promising potential biomarkers for the diagnosis of IPF. Our results demonstrate that individual lipid molecules have the ability to differentiate IPF from controls. Implications for future studies include validation of the accuracy of biomarkers to diagnose IPF and investigation in their IPF-specificity compared to other respiratory diseases, such as asthma, COPD, and infective pneumonia.

Acknowledgements

Not applicable.

Funding

Not applicable.

Availability of data and materials

The datasets supporting the conclusions of this article is available in the results section.
The clinical IPF samples included in this study were collected from the First hospital of Tsinghua University. All patient data were anonymous, so informed consent for participation was not required. The use of these samples was approved by the Institutional Review Board for human studies at the First Hospital of Tsinghua University.
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 Joo S, et al. Clinical results of sublobar resection versus lobectomy or more extensive resection for lung cancer patients with idiopathic pulmonary fibrosis. J Thorac Dis. 2016;8(5):977–84.CrossRefPubMedPubMedCentral Joo S, et al. Clinical results of sublobar resection versus lobectomy or more extensive resection for lung cancer patients with idiopathic pulmonary fibrosis. J Thorac Dis. 2016;8(5):977–84.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Tomassetti S, et al. The impact of lung cancer on survival of idiopathic pulmonary fibrosis. Chest. 2015;147(1):157–64.CrossRefPubMed Tomassetti S, et al. The impact of lung cancer on survival of idiopathic pulmonary fibrosis. Chest. 2015;147(1):157–64.CrossRefPubMed
3.
Zurück zum Zitat Ghatol A, Ruhl AP, Danoff SK. Exacerbations in idiopathic pulmonary fibrosis triggered by pulmonary and nonpulmonary surgery: a case series and comprehensive review of the literature. Lung. 2012;190(4):373–80.CrossRefPubMed Ghatol A, Ruhl AP, Danoff SK. Exacerbations in idiopathic pulmonary fibrosis triggered by pulmonary and nonpulmonary surgery: a case series and comprehensive review of the literature. Lung. 2012;190(4):373–80.CrossRefPubMed
4.
Zurück zum Zitat King TE Jr, Pardo A, Selman M. Idiopathic pulmonary fibrosis. Lancet. 2011;378(9807):1949–61.CrossRefPubMed King TE Jr, Pardo A, Selman M. Idiopathic pulmonary fibrosis. Lancet. 2011;378(9807):1949–61.CrossRefPubMed
5.
Zurück zum Zitat Chuchalin AG. Idiopathic pulmonary fibrosis. Ter Arkh. 2000;72(3):5–12.PubMed Chuchalin AG. Idiopathic pulmonary fibrosis. Ter Arkh. 2000;72(3):5–12.PubMed
6.
Zurück zum Zitat Khalil W, et al. Pathologic regulation of collagen I by an aberrant protein Phosphatase 2A/Histone Deacetylase C4/MicroRNA-29 signal Axis in idiopathic pulmonary fibrosis fibroblasts. Am J Respir Cell Mol Biol. 2015;53(3):391–9.CrossRefPubMedPubMedCentral Khalil W, et al. Pathologic regulation of collagen I by an aberrant protein Phosphatase 2A/Histone Deacetylase C4/MicroRNA-29 signal Axis in idiopathic pulmonary fibrosis fibroblasts. Am J Respir Cell Mol Biol. 2015;53(3):391–9.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Romero Y, et al. mTORC1 activation decreases autophagy in aging and idiopathic pulmonary fibrosis and contributes to apoptosis resistance in IPF fibroblasts. Aging Cell. 2016;15(6):1103–12.CrossRef Romero Y, et al. mTORC1 activation decreases autophagy in aging and idiopathic pulmonary fibrosis and contributes to apoptosis resistance in IPF fibroblasts. Aging Cell. 2016;15(6):1103–12.CrossRef
9.
Zurück zum Zitat Zhao YY, Cheng XL, Lin RC. Lipidomics applications for discovering biomarkers of diseases in clinical chemistry. Int Rev Cell Mol Biol. 2014;313:1–26.CrossRefPubMed Zhao YY, Cheng XL, Lin RC. Lipidomics applications for discovering biomarkers of diseases in clinical chemistry. Int Rev Cell Mol Biol. 2014;313:1–26.CrossRefPubMed
10.
11.
Zurück zum Zitat Zehethofer N, et al. Lipid analysis of airway epithelial cells for studying respiratory diseases. Chromatographia. 2015;78(5–6):403–13.CrossRefPubMed Zehethofer N, et al. Lipid analysis of airway epithelial cells for studying respiratory diseases. Chromatographia. 2015;78(5–6):403–13.CrossRefPubMed
12.
Zurück zum Zitat Simons K, Toomre D. Lipid rafts and signal transduction. Nat Rev Mol Cell Biol. 2000;1(1):31–9.CrossRefPubMed Simons K, Toomre D. Lipid rafts and signal transduction. Nat Rev Mol Cell Biol. 2000;1(1):31–9.CrossRefPubMed
13.
Zurück zum Zitat Farooqui AA. Lipid mediators in the neural cell nucleus: their metabolism, signaling, and association with neurological disorders. Neuroscientist. 2009;15(4):392–407.CrossRefPubMed Farooqui AA. Lipid mediators in the neural cell nucleus: their metabolism, signaling, and association with neurological disorders. Neuroscientist. 2009;15(4):392–407.CrossRefPubMed
15.
Zurück zum Zitat Wood PL. Mass spectrometry strategies for clinical metabolomics and lipidomics in psychiatry, neurology, and neuro-oncology. Neuropsychopharmacology. 2014;39(1):24–33.CrossRefPubMed Wood PL. Mass spectrometry strategies for clinical metabolomics and lipidomics in psychiatry, neurology, and neuro-oncology. Neuropsychopharmacology. 2014;39(1):24–33.CrossRefPubMed
16.
Zurück zum Zitat Min HK, Lim S, Chung BC, Moon MH. Shotgun lipidomics for candidate biomarkers of urinary phospholipids in prostate cancer. Anal Bioanal Chem. 2011;399(2):823–30.CrossRefPubMed Min HK, Lim S, Chung BC, Moon MH. Shotgun lipidomics for candidate biomarkers of urinary phospholipids in prostate cancer. Anal Bioanal Chem. 2011;399(2):823–30.CrossRefPubMed
17.
Zurück zum Zitat Kim IC, et al. Lipid profiles for HER2-positive breast cancer. Anticancer Res. 2013;33(6):2467–72.PubMed Kim IC, et al. Lipid profiles for HER2-positive breast cancer. Anticancer Res. 2013;33(6):2467–72.PubMed
18.
19.
Zurück zum Zitat de Gomez Dumm NT, Giammona AM, Touceda LA, Raimondi C. Lipid abnormalities in chronic renal failure patients undergoing hemodialysis. Medicina (B Aires). 2001;61(2):142–6. de Gomez Dumm NT, Giammona AM, Touceda LA, Raimondi C. Lipid abnormalities in chronic renal failure patients undergoing hemodialysis. Medicina (B Aires). 2001;61(2):142–6.
20.
Zurück zum Zitat Yang Y, Uhlig S. The role of sphingolipids in respiratory disease. Ther Adv Respir Dis. 2011;5(5):325–44.CrossRefPubMed Yang Y, Uhlig S. The role of sphingolipids in respiratory disease. Ther Adv Respir Dis. 2011;5(5):325–44.CrossRefPubMed
21.
22.
Zurück zum Zitat Masini E, et al. Ceramide: a key signaling molecule in a Guinea pig model of allergic asthmatic response and airway inflammation. J Pharmacol Exp Ther. 2008;324(2):548–57.CrossRefPubMed Masini E, et al. Ceramide: a key signaling molecule in a Guinea pig model of allergic asthmatic response and airway inflammation. J Pharmacol Exp Ther. 2008;324(2):548–57.CrossRefPubMed
23.
Zurück zum Zitat Teichgraber V, et al. Ceramide accumulation mediates inflammation, cell death and infection susceptibility in cystic fibrosis. Nat Med. 2008;14(4):382–91.CrossRefPubMed Teichgraber V, et al. Ceramide accumulation mediates inflammation, cell death and infection susceptibility in cystic fibrosis. Nat Med. 2008;14(4):382–91.CrossRefPubMed
24.
Zurück zum Zitat Ley B, Brown KK, Collard HR. Molecular biomarkers in idiopathic pulmonary fibrosis. American journal of physiology. Lung Cell Mol Physiol. 2014;307(9):L681–91.CrossRef Ley B, Brown KK, Collard HR. Molecular biomarkers in idiopathic pulmonary fibrosis. American journal of physiology. Lung Cell Mol Physiol. 2014;307(9):L681–91.CrossRef
25.
26.
Zurück zum Zitat Schwudke D, Schuhmann K, Herzog R, Bornstein SR, Shevchenko A. Shotgun lipidomics on high resolution mass spectrometers. Cold Spring Harb Perspect Biol. 2011;3(9):a004614.CrossRefPubMedPubMedCentral Schwudke D, Schuhmann K, Herzog R, Bornstein SR, Shevchenko A. Shotgun lipidomics on high resolution mass spectrometers. Cold Spring Harb Perspect Biol. 2011;3(9):a004614.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Yang K, Cheng H, Gross RW, Han X. Automated lipid identification and quantification by multidimensional mass spectrometry-based shotgun lipidomics. Anal Chem. 2009;81(11):4356–68.CrossRefPubMedPubMedCentral Yang K, Cheng H, Gross RW, Han X. Automated lipid identification and quantification by multidimensional mass spectrometry-based shotgun lipidomics. Anal Chem. 2009;81(11):4356–68.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Southam AD, et al. Drug redeployment to kill leukemia and lymphoma cells by disrupting SCD1-mediated synthesis of monounsaturated fatty acids. Cancer Res. 2015;75(12):2530–40.CrossRefPubMed Southam AD, et al. Drug redeployment to kill leukemia and lymphoma cells by disrupting SCD1-mediated synthesis of monounsaturated fatty acids. Cancer Res. 2015;75(12):2530–40.CrossRefPubMed
29.
Zurück zum Zitat McLaren DG, et al. An ultraperformance liquid chromatography method for the normal-phase separation of lipids. Anal Biochem. 2011;414(2):266–72.CrossRefPubMed McLaren DG, et al. An ultraperformance liquid chromatography method for the normal-phase separation of lipids. Anal Biochem. 2011;414(2):266–72.CrossRefPubMed
31.
Zurück zum Zitat Kuhn T, et al. Higher plasma levels of lysophosphatidylcholine 18:0 are related to a lower risk of common cancers in a prospective metabolomics study. BMC Med. 2016;14:13.CrossRefPubMedPubMedCentral Kuhn T, et al. Higher plasma levels of lysophosphatidylcholine 18:0 are related to a lower risk of common cancers in a prospective metabolomics study. BMC Med. 2016;14:13.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Piszcz J, et al. To treat or not to treat: metabolomics reveals biomarkers for treatment indication in chronic lymphocytic leukaemia patients. Oncotarget. 2016;7(16):22324–38.CrossRefPubMedPubMedCentral Piszcz J, et al. To treat or not to treat: metabolomics reveals biomarkers for treatment indication in chronic lymphocytic leukaemia patients. Oncotarget. 2016;7(16):22324–38.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Zhao YY, Lin RC. UPLC-MS(E) application in disease biomarker discovery: the discoveries in proteomics to metabolomics. Chem Biol Interact. 2014;215:7–16.CrossRefPubMed Zhao YY, Lin RC. UPLC-MS(E) application in disease biomarker discovery: the discoveries in proteomics to metabolomics. Chem Biol Interact. 2014;215:7–16.CrossRefPubMed
34.
Zurück zum Zitat Wilson ID, et al. High resolution "ultra performance" liquid chromatography coupled to oa-TOF mass spectrometry as a tool for differential metabolic pathway profiling in functional genomic studies. J Proteome Res. 2005;4(2):591–8.CrossRefPubMed Wilson ID, et al. High resolution "ultra performance" liquid chromatography coupled to oa-TOF mass spectrometry as a tool for differential metabolic pathway profiling in functional genomic studies. J Proteome Res. 2005;4(2):591–8.CrossRefPubMed
35.
Zurück zum Zitat Wang CZ, et al. Ultra-performance liquid chromatography and time-of-flight mass spectrometry analysis of ginsenoside metabolites in human plasma. Am J Chin Med. 2011;39(6):1161–71.CrossRefPubMedPubMedCentral Wang CZ, et al. Ultra-performance liquid chromatography and time-of-flight mass spectrometry analysis of ginsenoside metabolites in human plasma. Am J Chin Med. 2011;39(6):1161–71.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Raghu G, 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(6):788–824.CrossRefPubMedPubMedCentral Raghu G, 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(6):788–824.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Pedersen HK, et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature. 2016;535(7612):376–81.CrossRefPubMed Pedersen HK, et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature. 2016;535(7612):376–81.CrossRefPubMed
38.
Zurück zum Zitat Fahy E, et al. A comprehensive classification system for lipids. J Lipid Res. 2005;46(5):839–61.CrossRefPubMed Fahy E, et al. A comprehensive classification system for lipids. J Lipid Res. 2005;46(5):839–61.CrossRefPubMed
39.
Zurück zum Zitat Ohnishi H, 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(3):378–81.CrossRefPubMed Ohnishi H, 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(3):378–81.CrossRefPubMed
40.
Zurück zum Zitat Oikonomidi S, et al. Matrix metalloproteinases in respiratory diseases: from pathogenesis to potential clinical implications. Curr Med Chem. 2009;16(10):1214–28.CrossRefPubMed Oikonomidi S, et al. Matrix metalloproteinases in respiratory diseases: from pathogenesis to potential clinical implications. Curr Med Chem. 2009;16(10):1214–28.CrossRefPubMed
41.
Zurück zum Zitat Ollero M, et al. Plasma lipidomics reveals potential prognostic signatures within a cohort of cystic fibrosis patients. J Lipid Res. 2011;52(5):1011–22.CrossRefPubMedPubMedCentral Ollero M, et al. Plasma lipidomics reveals potential prognostic signatures within a cohort of cystic fibrosis patients. J Lipid Res. 2011;52(5):1011–22.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Goss V, Hunt AN, Postle AD. Regulation of lung surfactant phospholipid synthesis and metabolism. Biochim Biophys Acta. 2013;1831(2):448–58.CrossRefPubMed Goss V, Hunt AN, Postle AD. Regulation of lung surfactant phospholipid synthesis and metabolism. Biochim Biophys Acta. 2013;1831(2):448–58.CrossRefPubMed
43.
Zurück zum Zitat Frisardi V, Panza F, Seripa D, Farooqui T, Farooqui AA. Glycerophospholipids and glycerophospholipid-derived lipid mediators: a complex meshwork in Alzheimer's disease pathology. Prog Lipid Res. 2011;50(4):313–30.CrossRefPubMed Frisardi V, Panza F, Seripa D, Farooqui T, Farooqui AA. Glycerophospholipids and glycerophospholipid-derived lipid mediators: a complex meshwork in Alzheimer's disease pathology. Prog Lipid Res. 2011;50(4):313–30.CrossRefPubMed
44.
Zurück zum Zitat Guerrera IC, et al. A novel lipidomic strategy reveals plasma phospholipid signatures associated with respiratory disease severity in cystic fibrosis patients. PLoS One. 2009;4(11):e7735.CrossRefPubMedPubMedCentral Guerrera IC, et al. A novel lipidomic strategy reveals plasma phospholipid signatures associated with respiratory disease severity in cystic fibrosis patients. PLoS One. 2009;4(11):e7735.CrossRefPubMedPubMedCentral
45.
46.
Zurück zum Zitat Wanders RJ, et al. Peroxisomal fatty acid alpha- and beta-oxidation in health and disease: new insights. Adv Exp Med Biol. 2003;544:293–302.CrossRefPubMed Wanders RJ, et al. Peroxisomal fatty acid alpha- and beta-oxidation in health and disease: new insights. Adv Exp Med Biol. 2003;544:293–302.CrossRefPubMed
47.
49.
50.
Zurück zum Zitat Scarpa MC, et al. Ceramide expression and cell homeostasis in chronic obstructive pulmonary disease. Respiration. 2013;85(4):342–9.CrossRefPubMed Scarpa MC, et al. Ceramide expression and cell homeostasis in chronic obstructive pulmonary disease. Respiration. 2013;85(4):342–9.CrossRefPubMed
51.
Zurück zum Zitat Miao H, et al. Plasma lipidomics reveal profound perturbation of glycerophospholipids, fatty acids, and sphingolipids in diet-induced hyperlipidemia. Chem Biol Interact. 2015;228:79–87.CrossRefPubMed Miao H, et al. Plasma lipidomics reveal profound perturbation of glycerophospholipids, fatty acids, and sphingolipids in diet-induced hyperlipidemia. Chem Biol Interact. 2015;228:79–87.CrossRefPubMed
52.
Zurück zum Zitat Ishikawa M, et al. Plasma and serum lipidomics of healthy white adults shows characteristic profiles by subjects' gender and age. PLoS One. 2014;9(3):e91806.CrossRefPubMedPubMedCentral Ishikawa M, et al. Plasma and serum lipidomics of healthy white adults shows characteristic profiles by subjects' gender and age. PLoS One. 2014;9(3):e91806.CrossRefPubMedPubMedCentral
53.
Metadaten
Titel
Identification of the lipid biomarkers from plasma in idiopathic pulmonary fibrosis by Lipidomics
verfasst von
Feng Yan
Zhensong Wen
Rui Wang
Wenling Luo
Yufeng Du
Wenjun Wang
Xianyang Chen
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
BMC Pulmonary Medicine / Ausgabe 1/2017
Elektronische ISSN: 1471-2466
DOI
https://doi.org/10.1186/s12890-017-0513-4

Weitere Artikel der Ausgabe 1/2017

BMC Pulmonary Medicine 1/2017 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Neu im Fachgebiet Innere Medizin

22.04.2024 | DGIM 2024 | Kongressbericht | Nachrichten

Krebspatienten impfen: Was? Wen? Und wann nicht?

22.04.2024 | DGIM 2024 | Kongressbericht | Nachrichten

Nierenultraschall: Tipps vom Profi

22.04.2024 | DGIM 2024 | Kongressbericht | Nachrichten

„KI sieht, was wir nicht sehen“

Update Innere Medizin

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