Skip to main content
Erschienen in: Respiratory Research 1/2015

Open Access 01.12.2015 | Research

Genome-wide mRNA expression profiling in vastus lateralis of COPD patients with low and normal fat free mass index and healthy controls

verfasst von: Roberto A Rabinovich, Ellen Drost, Jonathan R Manning, Donald R Dunbar, MaCarmen Díaz-Ramos, Ramzi Lakhdar, Ricardo Bastos, William MacNee

Erschienen in: Respiratory Research | Ausgabe 1/2015

Abstract

Background

Chronic Obstructive Pulmonary Disease (COPD) has significant systemic effects beyond the lungs amongst which muscle wasting is a prominent contributor to exercise limitation and an independent predictor of morbidity and mortality. The molecular mechanisms leading to skeletal muscle dysfunction/wasting are not fully understood and are likely to be multi-factorial. The need to develop therapeutic strategies aimed at improving skeletal muscle dysfunction/wasting requires a better understanding of the molecular mechanisms responsible for these abnormalities. Microarrays are powerful tools that allow the investigation of the expression of thousands of genes, virtually the whole genome, simultaneously. We aim at identifying genes and molecular pathways involved in skeletal muscle wasting in COPD.

Methods

We assessed and compared the vastus lateralis transcriptome of COPD patients with low fat free mass index (FFMI) as a surrogate of muscle mass (COPDL) (FEV1 30 ± 3.6%pred, FFMI 15 ± 0.2 Kg.m−2) with patients with COPD and normal FFMI (COPDN) (FEV1 44 ± 5.8%pred, FFMI 19 ± 0.5 Kg.m−2) and a group of age and sex matched healthy controls (C) (FEV1 95 ± 3.9%pred, FFMI 20 ± 0.8 Kg.m−2) using Agilent Human Whole Genome 4x44K microarrays. The altered expression of several of these genes was confirmed by real time TaqMan PCR. Protein levels of P21 were assessed by immunoblotting.

Results

A subset of 42 genes was differentially expressed in COPDL in comparison to both COPDN and C (PFP < 0.05; −1.5 ≥ FC ≥ 1.5). The altered expression of several of these genes was confirmed by real time TaqMan PCR and correlated with different functional and structural muscle parameters. Five of these genes (CDKN1A, GADD45A, PMP22, BEX2, CGREF1, CYR61), were associated with cell cycle arrest and growth regulation and had been previously identified in studies relating muscle wasting and ageing. Protein levels of CDKN1A, a recognized marker of premature ageing/cell cycle arrest, were also found to be increased in COPDL.

Conclusions

This study provides evidence of differentially expressed genes in peripheral muscle in COPD patients corresponding to relevant biological processes associated with skeletal muscle wasting and provides potential targets for future therapeutic interventions to prevent loss of muscle function and mass in COPD.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12931-014-0139-5) contains supplementary material, which is available to authorized users.
Ricardo Bastos and William MacNee contributed equally to this work.

Competing interests

The authors have no competing interests to declare. This study was financially supported by the Chief Scientist Office (CSO 06/S1103/5) and the Fondo de Investigación Sanitaria (FIS) (PI08/0320).

Authors’ contributions

Conception and design: RR, ED, RB, WM. Analysis and interpretation: RR, ED, JM, DD, MCDR, RB, WM. Drafting the manuscript for important intellectual contents: RR, ED, RB, WM. All authors read and approved the final manuscript.
Abkürzungen
mRNA
m Ribonucleic acid
COPD
Chronic obstructive pulmonary disease
FFMI
Fat free mass index
COPDL
Patients with COPD and low FFMI
COPDN
Patients with COPD and normal FFMI
C
Control subjects
HRQoL
Health related quality of life
BIA
Bioimpedance analysis
6MWD
Six minute walking distance
QMVC
Quadriceps maximal voluntary contraction
SGRQ
Saint George’s respiratory questionnaire
mMRC
Modified medical research council dyspnoea score
PA
Physical activity
PAV
Physical activity assessed with the Voorrips questionnaire
LCADL
London chest activity of daily living scale
PAL
Physical activity assessed with the LCADL
PFP
Percent false positives
FDR
False discovery rate
GO
Gene ontology
qPCR
Real time polymerase chain reaction
ΔCT
Delta cycle threshold
ADL
Activities of daily living
FEV1
Forced expiratory volume in the first second
FVC
Forced vital capacity
RP
Rank products
DEGs
Differentially expressed genes
FC
Fold change
DNA
Deoxyribonucleic acid
ICU
Intensive care unit
PaO2
Arterial oxygen partial pressure
PaCO2
Arterial carbon dioxide partial pressure

Introduction

Chronic obstructive pulmonary disease (COPD) is associated with several extra-pulmonary effects of which skeletal muscle wasting is one of the most extensively studied [1,2] and results in loss of muscle strength [1,3-6], contributes to exercise (in)tolerance [7-10] and is a predictor of health related quality of life (HRQoL) [11] and survival [12,13] independent of the degree of airway obstruction [10].
Muscle wasting affects 18 to 36% of patients with COPD [7,14] and can be present even in patients with normal weight [7,14,15]. Indeed, muscle wasting is a better predictor of health related quality of life [11] and survival [12,13] than body weight itself. But why does only a subgroup of patients with COPD develop muscle wasting?; Several patho-physiological changes, have been identified in the skeletal muscle of COPD patients namely fibre size reduction (atrophy) [7], fibre type redistribution [16], altered bioenergetics [16], altered capillarization [17], and altered mitochondrial function [18,19].
The molecular mechanisms leading to skeletal muscle wasting are not fully understood and are likely to be multi-factorial, including physical inactivity, systemic inflammation/oxidative stress and cell hypoxia [1] among others.
Accumulating evidence supports the idea that COPD is a disease of accelerated ageing [20]. It has recently been shown that limb muscles of patients with COPD have increased number of senescent satellite cells and an exhausted muscle regenerative capacity, compromising the maintenance of muscle mass in these individuals [21] suggesting that premature cellular senescence and subsequent exhaustion of the regenerative potential of the muscles may relate to muscle abnormalities characteristic of these patients.
Strategies to reverse skeletal muscle dysfunction/wasting achieve only relatively modest improvements [22]. There is a need to develop therapeutic strategies aimed at improving skeletal muscle dysfunction/wasting, which requires a better understanding of the molecular mechanisms responsible for these abnormalities.
Microarrays are powerful tools that allow the investigation of the expression of thousands of genes, virtually the whole genome, simultaneously. An analysis of the genes that are being transcribed in the muscle, the transcriptome, should shed light on the molecular mechanisms responsible for muscle dysfunction and wasting in COPD and can help to identify molecular targets for the development of therapeutic strategies specifically designed to improve muscle function and bulk.
In this study we assessed the transcriptome of the vastus lateralis muscle in COPD patients with low fat free mass index (FFMI) as a surrogate of muscle mass (COPDL) in comparison to patients with COPD and normal FFMI (COPDN) and a group of age and sex matched healthy controls (C).
We hypothesize that genes related to cell cycle arrest and inhibition of cell growth will be up-regulated while genes related to energy production and muscle development will be down-regulated in COPDL. We expect similarities in the transcriptome of COPDL and muscle wasting relating to the normal ageing process. Moreover, the transcriptome analysis of this group may reveal important pathways leading to peripheral muscle wasting.
This study demonstrates that vastus lateralis of patients with COPD and muscle wasting overexpress genes related to inhibition of cell cycle and of cell growth whilst genes related to muscle formation and growth and energy production were down-regulated. This pattern is similar to observations associated with ageing, which suggests that premature ageing may play a role in muscle atrophy in COPD.

Methods

Study group

Nineteen stable patients with COPD, nine with low FFMI (COPDL) and ten with normal FFMI (COPDN), and ten age, gender and smoking status-matched healthy subjects with normal FFMI were included in the present study (Table 1). All patients had a diagnosis of COPD according to the Global Initiative for Chronic Obstructive Lung Disease [23]. They were clinically stable and free of exacerbations for 4 weeks prior to the study and free of drugs that can potentially affect the muscle (i.e. systemic corticosteroids, statins). The study was approved by the Lothian Regional Ethics Committee.
Table 1
Characteristics of the study groups
 
COPD L
COPD N
Controls
p-value
M/F
7/2
A
8/2
A
8/2
A
ns
Age (Years)
67 ± 2.0
A
69 ± 1.5
A
68 ± 1.4
A
ns
BMI (Kg.m−2)
18.8 ± 0.7
A
26 ± 0.7
B
30 ± 2.1
C
<0.0001
FFMI (Kg.m−2)
15 ± 0.2
A
19 ± 0.5
B
20 ± 0.8
B
<0.0001
Active/ex-smokers
1/8
A
2/8
A
2/8
A
ns
Pack/year
66 ± 14
A
49 ± 6.6
AB
32 ± 5.0
B
0.037
Average cessation (years)
6.0 ± 2.5
A
8.4 ± 2.2
A
23 ± 5.5
B
<0.01
Age at smoking cessation (years)
61.1 ± 8.2
A
60.5 ± 7.6
A
45.1 ± 15.5
B
<0.01
mMRC
4 ± 0.4
A
3 ± 0.4
B
  
0.038
FEV1 (L)
0.8 ± 0.1
A
1.2 ± 0.1
A
2.8 ± 0.2
B
<0.0001
FEV1 (% pred)
30 ± 3.6
A
44 ± 5.8
A
95 ± 3.9
B
<0.0001
FVC (L)
2.6 ± 0.3
A
2.7 ± 0.4
A
3.9 ± 0.2
B
0.015
FVC (% pred)
76 ± 6.4
A
88 ± 9.0
AB
104 ± 3.2
B
0.01
FEV1/FVC
0.32 ± 0.1
A
0.38 ± 0.1
A
0.71 ± 0.0
B
<0.0001
PaO2 (mmHg)
79 ± 7.4
A
70 ± 3.1
A
73 ± 1.8
A
ns
PaCO2 (mmHg)
43 ± 2.0
A
41 ± 1.1
A
42 ± 0.6
A
ns
Physical activity (V)
1.2 ± 0.3
A
7 ± 1.9
B
11 ± 1.5
B
<0.0001
Physical activity (L)
43 ± 3.6
A
30 ± 5.0
A
  
ns
QMVC (N)
213 ± 19.4
A
323 ± 25.6
B
366 ± 26.8
B
0.002
6MWD (m)
306 ± 55
A
411 ± 50.1
AB
524 ± 37.9
B
0.01
BODE
6.5 ± 1.3
A
4 ± 0.9
A
  
ns
SGRQ Symptoms
76 ± 4.8
A
60 ± 4.2
B
  
0.01
SGRQ Activity
86 ± 4.7
A
52 ± 9.2
B
  
0.003
SGRQ Impact
57 ± 6.6
A
33 ± 7.7
B
  
0.01
SGRQ Total
69 ± 5.4
A
43 ± 6.8
B
  
0.005
Type I Fibre (%)
25.1 ± 4.6
A
24.4 ± 3.3
A
38.7 ± 3.9
B
0.04
Type II area (μ2)
2033 ± 166
A
2978 ± 277
B
2564 ± 277
AB
0.03
Definition of abbreviations: COPDN = COPD patients with normal FFMI; COPDL = patients with COPD with low FFMI; BMI = Body mass index; FFMI = fat free mass index; MRC = medical research council dyspnoea score; FEV1 = forced expiratory volume in the first second; FVC = forced vital capacity; PaO2 = arterial oxygen partial pressure; PaCO2 = arterial carbon dioxide partial pressure; Physical Activity (V) = Voorrips Questionnaire; Physical activity (L) = London Chest Activity of Daily Living Scale; QMVC = quadriceps maximal voluntary contraction; 6MWD = six minute walking distance; SGRQ = St. George’s Respiratory Questionnaire; ns = not significant; NA = not applicable. Comparisons among groups were done using ANOVA and Student-Newman-Keuls as a post-hoc test. Differences among the three different groups were stated using letters A, B and C where sharing a letter implies no differences between these groups and having a different letter implies a statistical difference in the post-hoc test.

Measurements

Assessment

All subjects had the following baseline assessments: anthropometric measurements, body composition measurement with bioimpedance (BIA), pulmonary function tests (spirometry) and blood gases (Ciba Corning 800, USA), six–minute walking distance (6MWD) [24], quadriceps maximal voluntary contraction (QMVC) [25] (Chatillon® K-MSC 500, Ametek, Florida), health-related quality of life questionnaires (St. George’s Respiratory Questionnaire, SGRQ) [26], modified Medical Research Council (mMRC) dyspnoea scale and physical activity (PA) levels using the Voorrips physical activity questionnaire (PAV) [27] and the London Chest Activity of Daily Living Scale (LCADL) [28] for patients with COPD. Number of exacerbations in the previous year was recorded.
Low FFMI was defined as <16 kg.m−2 for male and <15 kg.m−2 for female COPD patients [29].

Vastus Lateralis muscle biopsy and RNA isolation

An open muscle biopsy of the “vastus lateralis” was obtained and ~0.1 g was included in RNA stabilization reagent (RNAlater®, Ambion, Inc., USA) and stored at −20°C for RNA extraction. Total RNA was extracted and purified by homogenisation (TissueLyser, Qiagen Ltd. West Sussex, UK) of tissue employing the TRIzol® Plus RNA Purification Kit (Invitrogen Life Technologies, Carlsbad, CA).

Fibre type typification

Paraffin sections (5um) were de-waxed and re-hydrated through graded ethanol using standard procedures. Sections were placed in 250 ml of Novocastra pH8 retrieval buffer and subjected to antigen retrieval in a de-cloaking chamber (Biocare Medical, USA) using a protocol described elsewhere [30].

Microarray hybridization and data analysis

Five hundred nanograms of total RNA from each sample was converted into labelled cRNA with nucleotides coupled to a fluorescent dye (Cy3) using the Quick Amp Kit (Agilent Technologies, Palo Alto, CA). Cy3-labeled cRNA (1.65 μg) was hybridized to Agilent Human Whole Genome 4×44K Microarrays (Agilent Technologies, Santa Clara, CA). The hybridized array was then washed and scanned and the data were extracted from the scanned image using Feature Extraction version 10.2 (Agilent Technologies).
Pre-processing (background correction, normalization, filtering and summarization) subsequent data processing and analysis was performed using the Agi4x44 Pre-process module from Bioconductor [31,32].
The Rank Product [33,34] was employed for the microarray data analysis. RP is a non-parametric algorithm that detects probes/genes consistently highly ranked by fold-change between samples from different groups and employs a ‘percent false positives’ (PFP) measure, also known as false discovery rate (FDR), to select the most significant differential expressions. A percentage of false positive (PFP) below 0.05 was considered statistically significant.
The gene functional enrichment analysis was performed using DAVID Bioinformatics Resources (http://​david.​abcc.​ncifcrf.​gov/​). Specifically, the Functional Annotation Chart tool was used to enrich the over-represented Gene Ontology (GO) terms among the differentially expressed gene list. A list of all detectable transcripts was used as the background for the GO analysis [35]. The GO terms after correction for FDR at p < 0.05 (Benjamini Hochberg) were selected for further analysis and interpretation.
Ingenuity Pathway Analysis (IPA, Ingenuity Systems, Redwood, CA) was used to further investigate the Genespring expression clusters [36].

qPCR validation

Based on microarray-derived fold-change (>2) or statistical significance for differential expression and/or the biological relevance for the different comparisons, 11 genes were selected for TaqMan qPCR validation (Applied Biosystems).

Western blot analysis

P21, the protein encoded by the CDKN1A gene was determined using immunoblotting. 20 μg of protein was resolved by sodium-dodecyl sulfate-polyacrylamide gel electrophoresis on 10% polyacrylamide gels. Proteins were transferred to Immobilon-P PVDF membranes (Millipore, Billerica, MA), blocked with 5% dry milk (Bio-Rad, München, Germany) in TBS (Sigma) overnight at 4°C and probed with primary antibodies primary antibody against p21 (ab 7960) (Abcam, Bristol, UK) during 1 h at room temperature. Proteins were then visualized using the ECL Detection System (Pierce, Rockford, IL) as per the manufacturer’s instructions.

Statistical analysis

Anthropometric, physiological data and immunobloting results for p21 are expressed as mean ± SEM. These data were analysed using ANOVA with Student-Newman-Keuls as a post-hoc test. Correlation analysis between variables was conducted using Pearson’s correlation index for continuous variables and Spearman’s correlation index for categorical variables. For the qPCR validation analysis differential expression analysis on individual sample values of ΔCT using Kruskal Wallis with a Nemenyi-Damico-Wolfe-Dunn post-hoc test was performed.
The statistics were conducted using the statistical package SAS version 9.3 (SAS Institute Inc, Cary, NC, USA). A p value <0.05 was taken as statistically significant.
Full details of the methods can be found in the Additional file 1.

Results

Anthropometric characteristics and pulmonary function data of study subjects are depicted in Table 1. Both groups of COPD patients showed airflow limitation compared to healthy controls (C) who all had normal spirometry, but there were no differences in spirometry between COPDN and COPDL (Table 1 and Additional file 2: Figure S1). Patients were matched by smoking status (two COPDN, one COPDL patient and two healthy controls were active smokers at the time of inclusion, p = ns). All subjects, including healthy controls, were exposed (current or ex-smokers) to cigarette smoking (no never-smokers were included in the study). Moreover, no differences in the number of years of smoking cessation was seen between COPDL (6.0 ± 2.5 years) and COPDN (8.4 ± 2.2 years) nor in the age of the patients when they stopped smoking (61.1 ± 8.2 years old) COPDL and (60.5 ± 7.6 years) COPDN. Both COPD groups showed less years of smoking cessation than controls (23.2 ± 5.5 years) (ANOVA P < 0.01).
Compared to C, COPDN had lower BMI and physical activity and COPDL had very different anthropometric characteristics.
Compared to COPDN, COPDL had significantly lower BMI, FFM and FFMI, poorer HRQoL with higher values in all of the domains of the St. George’s respiratory questionnaire, higher mMRC score and worse muscle function as assessed by QMVC. They also had lower levels of physical activity measured by the Voorrips questionnaire (PAV) but no difference were seen in activities of daily living (ADL) assessed with the LCADL (PAL), specifically designed to assess ADL in patients with COPD (Table 1).
Both COPD groups showed a redistribution of muscle fibre type with a higher proportion of type II fibres and a lower proportion type I in comparison to healthy controls. Type II fibre area was significantly reduced in COPDL in comparison with COPDN (Table 1).
In the whole COPD population FFMI correlated with FEV1 (r = 0.51, p < 0.05), mMRC dyspnoea score (rho = −0.48, p < 0.05), QMVC (r = 0.76, p < 0.001), and physical activity (PAV) (r = 0.61, p < 0.01) while skeletal muscle function (QMVC) correlated with FFMI (r = 0.76, p < 0.0001), exacerbation rate (rho = −0.57, p < 0.05), 6MWD (r = 0.62, p < 0.01), physical activity (PAV) (r = 0.53, p < 0.05) and BODE index (rho = −0.66, p < 0.05).

Global assessment of gene expression

Hierarchical and k-means clustering were undertaken with normalized data. No pattern emerged from this analysis. We therefore performed three pair-wise class comparisons: COPDL vs. COPDN, COPDL vs. C and COPDN vs. C, employing Rank Products (RP) to detect differentially expressed genes (DEGs). First, a list of up- or down-regulated genes for each comparison was selected based on a PFP <0.05 (Table 2) (no FC criterion was applied). Comparisons of both groups COPD patients with C showed the most differentially expressed genes and the comparison between the COPD groups the least.
Table 2
DEGs between COPD L , COPD N and C
  
COPD L vs COPD N
COPD L vs C
COPD N vs C
Up regulated
Probes
954
1352
1050
Genes
531
830
724
Down regulated
Probes
1054
1631
1201
Genes
748
1152
799
DEGs between COPDL vs. COPDN, COPDL vs. C and COPDN vs C. Number of up and down regulated probes and genes with a PFP < 0.05 in all three comparisons.
In order to select the most relevant DEGs related to muscle wasting in COPD, we selected a list of DEGs between COPDL and COPDN. Furthermore, among this list, we selected those genes that were also differentially expressed between COPDL and C (Figure 1). This list of 1110 DEGs (1763 probes) (454 DEGs up-regulated and 656 DEGs down-regulated) was used to conduct a functional enrichment analysis. Table 3 displays the most characteristic GO biological processes (with 8 or more genes representing each class/category/pathway) that were enriched in this list. Significant GO classes from the list of up-regulated DEG correspond to functional terms related to protein synthesis, muscle organ development and striated muscle contraction while functional terms related to glucose metabolism, energy production, striated muscle development and striated muscle contraction were identified from the list of down-regulated genes.
Table 3
GO Terms associated with relevant genes related to muscle wasting
Source
GO terms
Fold enrichment
N o of gene in the term
FDR
Up-regulated genes
Traslational initiation
7.9
11
0.001
Traslational elongation
14.7
49
6.05E-42
Ribosomal biogenesis
4.2
17
0.004
rRNA processing
4.9
15
0.002
Muscle organ development
3.2
20
1.62E-05
Striated muscle contraction
6.5
10
1.55E-05
Down-regulated genes
Glucose metabolic process
3.3
21
0.008
Energy derivation by oxidation of organic compounds
5.8
37
4.86E-15
Electron transport chain
6.2
31
5.77E-13
ATP synthesis coupled electron transport
9.3
22
1.91E-12
Striated muscle tissue development
3.4
18
0.02
Muscle contraction
4.7
31
1.94E-09
GO Terms associated with DEGs between COPDL vs. COPDN and COPDL vs. C.
Functional enrichment analysis of 454 up-regulated and 656 down-regulated DEGs between COPDL vs. COPDN and COPDL vs. C (FDR < 0.05) with the corresponding GO term, the fold enrichment, number of genes involved in the term and the FDR (False discovery Rate).
Since some of these DEGs displayed marginal changes in gene expression, we applied a more restrictive criterion, namely a fold change cutoff of 1.5 (1.5 ≥ FC ≥ 1.5) in order to select more robust genes. Figure 2 shows the list of DEGs generated between COPDL and both other groups of subjects with normal muscle mass using this criterion. When the combined criteria of both statistical significance and fold change were applied the numbers of DEGs were notably reduced. Eighty three probes, representing 64 DEGs, were identified between COPDL and COPDN. Fifty six of these probes, representing 42 DEGs, were also differentially expressed between COPDL and C (Table 4 and 5). These 42 DEGs were selected for further analysis. Although this shorter list of genes was not suitable for a functional enrichment analysis, the biological significance of these genes was related with the significant functional categories identified in the list of 1110 DEGs originally obtained with a PFP < 0.05. Among the up-regulated genes (Table 4) from this list of 42 DEGs, we have identified genes related to cell cycle inhibition and inhibition of cell growth (CDKN1A, GADD45A, PMP22, BEX2, CGREF1, CYR61), inhibition of sarcomeric organization (HDGF), stress inducible factor (ATF3), inhibition of glucose metabolism (PDK4), lipid metabolism (LPL), aminoacid transportation (SLC22A3, SLC38A1), muscle repair (ABRA) and myogenesis (NNMT, ANKRD1). Among the down-regulated genes (Table 5) we have identified genes related to fatty acid synthesis (MCAT), muscle formation (CHRDL2, IRX4, PMEPA1), glucose endocytosis (RAB10) and gluconeogenesis (GPT).
Table 4
Up-regulated DEGs between COPD L vs. COPD N and COPD L vs. C with a FC over 1.5
PROBE
Gene symbol
COPD L vs. COPD N
COPD L vs. C
  
PFP
FC
PFP
FC
A_32_P200144
IGHG1
0.044
5.3
0.0177
9.4
A_23_P43979
IGLL5
0.0435
5.1
0.0133
7.9
A_24_P104980
IGLL5
0.0252
4.3
0.0098
6.5
A_23_P398566
 
0.0133
2.9
0.0491
1.7
A_23_P46429
CYR61
0
2.8
0
2.9
A_24_P370946
CYR61
0.0009
2.7
0.0013
2.7
A_23_P46426
CYR61
0.0008
2.5
0.0005
2.6
A_23_P161218
ANKRD1
0.0074
2.5
0.0067
1.9
A_24_P376707
HDGF
0.0402
2.5
0.0169
2.6
A_23_P19733
SLC22A3
0.0004
2.4
0
4.6
A_23_P34915
ATF3
0.0062
2.1
0.0011
3.4
A_32_P60459
OTUD1
0
2.0
0
2.6
A_32_P219135
 
0.012
2.0
0.0042
2.4
A_23_P166248
RCAN1
0.0162
1.9
0.0417
1.6
A_23_P22735
BEX2
0.0085
1.9
0.0003
3.2
A_23_P127584
NNMT
0
1.9
0
3.0
A_23_P100711
PMP22
0
1.8
0
2.2
A_23_P49338
TNFRSF12A
0.0022
1.8
0.0019
1.8
A_23_P363399
SLC38A1
0.0257
1.8
0.0027
3.0
A_23_P146233
LPL
0.0003
1.8
0.0022
1.5
A_24_P243749
PDK4
0
1.7
0
1.6
A_23_P403445
CGREF1
0.0353
1.7
0.0446
1.6
A_24_P193295
RAB15
0.0004
1.7
0.0002
1.9
A_23_P208540
LOC644482
0.0095
1.7
0.0026
1.8
A_24_P261734
SLC38A1
0.0148
1.7
0.0012
2.9
A_23_P408095
DSTN
0.0091
1.6
0.033
1.6
A_23_P418031
 
0.0133
1.6
0.0169
1.7
A_23_P314024
HLA-F
0.0075
1.6
0.0026
1.8
A_23_P166109
FLRT3
0.0419
1.6
0.0145
1.8
A_23_P59210
CDKN1A
0.0074
1.5
0.0014
2.2
A_23_P23221
GADD45A
0.0055
1.5
0.0016
1.7
A_32_P234459
HLA-H
0.0206
1.5
0.0103
1.7
A_23_P350295
 
0.0009
1.5
0.0003
1.7
A_24_P50489
 
0.0024
1.5
0.0008
1.5
A_23_P125109
 
0.0036
1.5
0.0002
1.7
A_23_P313482
ABRA
0.0003
1.5
0.0002
1.5
All probes are differentially expressed with a PFP (percentage of false positive) <0.05 and a FC (fold change); −1.5 ≥ FC ≥ 1.5.
Table 5
Down-regulated DEGs between COPD L vs. COPD N and COPD L vs. C with a FC over 1.5
PROBE
Gene symbol
COPD L vs. COPD N
COPD L vs. C
  
PFP
FC
PFP
FC
A_24_P401294
FLJ35934
0
−1.5
0
−1.5
A_24_P96961
SPSB1
0.0076
−1.5
0.0004
−2.2
A_24_P576591
 
0.0206
−1.5
0.0096
−1.5
A_24_P319675
RAB10
0.0105
−1.5
0
−2.8
A_23_P57089
PMEPA1
0.0056
−1.5
0.0005
−1.8
A_23_P146339
GPT
0.0013
−1.6
0.0007
−1.6
A_23_P308763
FARP1
0.0047
−1.6
0.0018
−1.7
A_24_P419028
MOP-1
0.0124
−1.6
0.0013
−1.8
A_23_P37856
HBA2
0.0003
−1.6
0
−2.4
A_23_P205355
SERPINA5
0.0377
−1.7
0.0031
−2.2
A_24_P413126
PMEPA1
0.0064
−1.7
0.0006
−1.9
A_24_P368943
EVX1
0.0006
−1.7
0.0002
−1.7
A_23_P26457
HBA2
0.0005
−1.8
0
−2.5
A_32_P163891
 
0.0344
−1.8
0.0104
−2.4
A_24_P75190
HBD
0.0003
−1.9
0
−3.1
A_24_P20795
IRX4
0.0055
−1.9
0.005
−1.9
A_23_P13548
CHRDL2
0.0139
−2.0
0.0452
−1.6
A_24_P412734
PRSS36
0.0474
−2.3
0.0039
−1.9
A_24_P237328
MCAT
0.0483
−2.4
0.0123
−1.8
A_24_P335092
SAA1
0.0207
−2.6
0.0003
3.8
All probes are differentially expressed with a PFP (percentage of false positive) < 0.05 and a FC (fold change); −1.5 ≥ FC ≥ 1.5.

Relationship with muscle structure and function

In order to identify bona fide muscle wasting associated genes we selected, from this subset of 42 DEGs, those that correlated with FFMI in the whole population including the healthy control. Ten out of twenty six of the up-regulated DEGs were negatively correlated with FFMI while nine out of the sixteen down-regulated DEGs were positively correlated with FFMI (Table 6).
Table 6
DEGs between COPD L and both COPD N and C that that varied with percentage of FFMI
Source
Probe
Gene symbol
rho
p
Up-regulated genes
A_23_P22735
BEX2
−0.55
0.0019
A_23_P100711
PMP22
−0.48
0.0085
A_23_P166109
FLRT3
−0.4
0.0327
A_24_P193295
RAB15
−0.44
0.0167
A_23_P46426
CYR61
−0.38
0.0391
A_24_P370946
CYR61
−0.39
0.0358
A_23_P46429
CYR61
−0.39
0.0337
A_24_P261734
SLC38A1
−0.47
0.0098
A_23_P363399
SLC38A1
−0.47
0.01
A_23_P19733
SLC22A3
−0.55
0.002
A_23_P49338
TNFRSF12A
−0.36
0.05
A_32_P219135
 
−0.45
0.015
A_23_P34915
ATF3
−0.36
0.055
A_23_P161218
ANKRD1
−0.47
0.0099
Down-regulated genes
A_23_P13548
CHRDL2
0.44
0.017
A_24_P20795
IRX4
0.36
0.056
A_24_P368943
EVX1
0.36
0.057
A_24_P413126
PMEPA1
0.52
0.0034
A_23_P57089
PMEPA1
0.57
0.0012
A_23_P205355
SERPINA5
0.5
0.0054
A_24_P419028
MOP-1
0.37
0.045
A_23_P146339
GPT
0.36
0.054
A_24_P319675
RAB10
0.64
0.0002
A_24_P401294
FLJ35934
0.44
0.0157
Significant associations between up-regulated and down-regulated DEGs and FFMI. rho: spearman correlation index.
Since COPDL were characterized by type II fibre atrophy in comparison with COPDN, we explored the correlations between these DEGs and type II fibre area. Three of the up-regulated DEGs (PMP22, CGREF1 and LPL) were negatively correlated with type II fibre area while four of the down-regulated DEGs (SAA1, CHRLD2, and FARP1) were positively correlated with type II fibre area (Additional file 3: Table S1). In turn, several of the up-regulated (ANKRD1, CDKN1A, GADD45A, ATF3, RAB15, ABRA and SLC22A3) and down-regulated (EVX1, PMEPA1, GPT, RAB10, SPSB1) DEGs in COPDL correlate with the proportion of Type II fibres (Additional file 4: Table S2). Moreover several up-regulated (negatively) and down-regulated (positively) DEGs correlated with QMVC, a measurement of muscle function (Additional file 5: Table S3).

qPCR validation

Eleven genes (CDKN1A, CEBPA, CYR61, EFCAB7, EGR1, HMOX1, PDE11A, SAA1, SLC22A3, SLC38A1 and SLC43A2) were selected for qPCR validation of the microarray experiment based on either a microarray-derived fold-change (>2) or statistical significance for differential expression and/or the biological relevance for the different comparisons (COPDN vs COPDL, COPDN vs C and COPDL vs C) (Figure 3A and Additional file 6: Figure S2). As shown in Table 7, the altered expression of all these genes was confirmed by real time TaqMan PCR.
Table 7
qPCR Validation of microarray experiment
Gene symbol
Microarray selection criteria
qPCR result
CDKN1A
PFP <0.005, FC 2.19 COPDL vs C
p < 0.005
CEBPA
PFP <0.005, FC 2.08 COPDL vs C
p < 0.005
CYR61
PFP <0.005, FC 2.6 COPDL vs C
p < 0.005
EFCAB7
PFP <0.005, FC 2.63 COPDL vs C
p < 0.005
EGR1
PFP <0.005, FC −2.11 COPDN vs C
p < 0.05
HMOX1
PFP <0.005, FC −2.31 COPDL vs C
p = 0.06
PFP <0.005, FC −2.26 COPDN vs C
p < 0.05
PDE11
PFP <0.005, FC −2.28 COPDL vs C
p < 0.05
SAA1
PFP <0.005, FC 3.8 COPDL vs C
p < 0.005
PFP <0.005, FC 10.1 COPDN vs C
ns
SLC22A3
PFP <0.005, FC 2.46 COPDL vs COPDN
ns
PFP <0.005, FC 4.62 COPDL vs C
p < 0.05
SLC38A1
PFP <0.005, FC 3.02 COPDL vs C
p < 0.005
SLC43A2
PFP <0.005, FC −2.1 COPDL vs C
p < 0.005
Comparative results of eleven selected genes for validation between microarrays and qPCR.
Figure 3A shows qPCR data for CDKN1A, one of the six genes representing the group of genes related to cell cycle inhibition and inhibition of cell growth from the list of 42 DEGs identified between COPDL and both other groups of subjects with normal muscle mass. CDKN1A mRNA expression was increased in COPDL compared to both COPDN and C (p < 0.005) (Graphics for the remaining eight genes are depicted in the Additional file 6: Figure S2). The expression of several of these genes assessed by qPCR correlated with parameters of muscle structure and function. CDKN1A and ANKRD1 correlated with percentage of Type II fibres (Figure 4), CEBPA, CYR61, EFCAB7, SLC22A3 and SLC38A1 correlated negatively with FFMI (Figure 5). In turn, CEBPA correlated positively (rho = 0.45, p < 0.05) while SLC43A2 correlated negatively (rho = −0.65, p < 0.0005) with the proportion of Type II fibres. Moreover, CDKN1A, CYR61, CEBPA, EFCAB7, SLC22A3 correlated negatively with QMVC, a measurement of muscle function (Additional file 7: Figure S3).

Immunobloting

As CDKN1A was differentially expressed between COPDL and COPDN but also between COPDL and C, with a fold change over 1.5, and it also correlated with several measurements of skeletal muscle structure and function and is consistently overexpressed in other conditions characterised by muscle wasting, we chose to explore the protein levels of p21 in our samples by immunoblotting. This showed that p21 is over expressed in COPDL in comparison to both COPDN and C (Figure 3B). However, only the comparison between COPDL and COPDN was statistically significant.

Discussion

This study shows relevant changes in gene expression in the vastus lateralis of COPD patients with skeletal muscle wasting. We identified several up-regulated genes associated with cell cycle arrest and growth regulation, and down-regulated genes associated with muscle formation and glucose metabolism in patients with low FFMI. These DEGs correlated with several parameters of muscle structure and function. The altered expression of 11 genes was confirmed by qPCR and the increased level in COPDL of P21, the protein encoded by CDKN1A, was also corroborated by immunoblotting.
Our initial approach revealed extensive changes in gene expression that allowed us to identify several biologically relevant GO terms related to protein synthesis, muscle contraction and muscle organ development (from the up-regulated list of genes in COPDL) and oxidative energy production, glucose metabolism and striated muscle contraction and myogenesis (from the down-regulated list of genes in COPDL). The concomitant activation and deregulation of different genes involved in myogenesis reveal the complex nature of the muscle wasting process in this population of patients. This, together with the activation of genes related to protein synthesis, show a potentially adaptive, yet clearly ineffective, response at maintaining muscle mass [37].
Using a more restrictive approach we were able to discard genes with potential marginal changes and identify a subset of more substantial DEGs. This group of DEGs fulfil several criteria: a) they were differentially expressed between COPDL and COPDN, b) showed a fold change ≥1.5 or ≤1.5 between COPDL and COPDN, c) were also differentially expressed between COPDL and C, b) showed a fold change ≥1.5 or ≤1.5 between COPDL and C. These criteria markedly reduced the list of DEGs but reinforced the relevance of these genes for the process of muscle wasting. This approach identified up-regulated DEGs related to the inhibition of cell growth and cell cycle and down-regulated genes related to myogenesis and glucose metabolism in COPDL. Several of these DEGs correlate with FFMI, parameters of muscle structure and function such as type II fibre size (the fibres showing atrophy in COPDL), the proportion of type II fibres and QMVC, indicating a potential role of these genes in the muscle wasting process.
Among these DEGs, GADD45A is induced by DNA damage [38] and stress (such as fasting or immobilization) [39,40]. GADD45A represses genes involved in anabolic signalling and energy production and induces pro-atrophy genes [39,40]. Interestingly, genes related to energy production by oxidation and glucose metabolic processes were down-regulated in our population of COPDL. GADD45A [41] and CDKN1A [42-44] have previously been identified in several conditions associated with muscle atrophy. Moreover, myostatin suppresses muscle cell growth via the transcriptional regulation of CDKN1A [45]. p21, the protein encoded by this gene, can inhibit apoptosis (when present in the cytosol by arresting the cell cycle allowing for DNA repair) or promote apoptosis (in the nucleus and interacting with other proteins) [38,46,47]. The interaction between p21 and growth arrest and DNA damage-inducible genes such as GADD45A results in apoptosis [38]. Furthermore, CDKN1A and GADD45A are also related to other muscular disorders such as amyotrophic lateral sclerosis [43]. ATF3, another of the DEGs up-regulated in COPDL, together with GADD45A and CDKN1A, is also associated with cell cycle arrest in response to DNA damage [48]. ATF3 is induced by stress and is related to cardiac contractility abnormalities [49] and muscular disorders such as amyotrophic lateral sclerosis [43]. ATF3 gene expression correlated negatively with FFMI and QMVC in our population. Another gene up-regulated in COPDL, ANKRD1, has been associated with up-regulation of p21, one of its downstream targets [50]. ANKRD1 is expressed in the muscle and migrates to the nucleus when the muscle is under stress. It has been shown to be up-regulated during muscle atrophy [51]. Evidence suggests that ANKRD1 per se cannot initiate atrophy. However, the observation that p21 is increased whenever ANDRD1 is increased in different models of muscle atrophy highlights the importance of this interaction in the muscle wasting process [50]. Moreover, a switch towards fast-twitch fibres has been reported in models of atrophy in association with up-regulation of ANKRD1 [50]. It is of note that ANKRD1 expression was negatively correlated with FFMI and QMVC and positively correlated with the proportion of Type II fibres in our COPDL, as were CDKN1A, GADD45A1 and ATF3. GADD45A1 also correlated negatively with muscle function measured as QMVC. CDKN1A has also been shown to interact with CEBPA [46] leading to cell arrest [38], and was up-regulated in COPDL and confirmed by qPCR. In fact, CEBPA inhibits cell proliferation by stabilising p21 [52] and protecting against its degradation [53]. CEBPA and CDKN1A gene expression assessed by qPCR correlated negatively with FFMI and QMVC in our population. The simultaneous up-regulation of these genes in COPDL together with the strong association with FFMI, and parameters of skeletal muscle structure and function suggest that cell cycle arrest followed by apoptosis play a role in the process of muscle wasting in this population in response to stress. The presence of muscle cell apoptosis in peripheral muscle of patients with COPD is still controversial. Some groups have reported increased cell apoptosis, assessed as DNA fragmentation [54,55], whereas others failed to report apoptosis, assessed as active caspase-3 [56]. It is interesting that, as mentioned previously, p21 has a dual function and can promote or inhibit apoptosis [38,46,47]. Apoptosis induced by p21 is distinct from that induced by other pro-apoptotic agents and does not involve activation of caspases [38]. In fact, while the carboxyl terminus interacts with molecules such as GADD45A or CEBPA, the N-terminus can interact with procaspase-3 to block activation of caspase-3 [57]. Hence, apoptosis induced by p21 is not affected by caspase inhibitors [38]. This may explain the controversy in the literature regarding muscle cell apoptosis in peripheral muscle of patients with COPD.
Other genes known to be overexpressed under stress situations and inflammation were also up-regulated in COPDL, such as CYR61 whose expression increases with exercise in humans [58-60]. CYR61 is required for the migration of myoblasts during the regeneration process [61] and mediates angiogenesis [60,62]. On the other hand, CY61 is increased in the muscle in models of malnutrition [63] and muscle denervation [64] and promotes cell migration and immobilise inflammatory cells in the site of inflammation and tissue repair [65]. Moreover, CYR61 gene expression assessed by both, microarray and qPCR, correlated negatively with FFM and QMVC in our population. In line with these findings, oxidative stress has been consistently shown in limb muscle of patients with COPD [66], particularly in patients with muscle wasting [67]. While local muscle oxidative stress is induced by exercise [67], the exercise-induced increase in antioxidant enzymes is attenuated or inhibited in the muscle of underweight patients with COPD [68,69]. HMOX1, a gene involved in the response to oxidants known to protect against cytotoxicity of oxidative stress and nitric oxide metabolism, was down-regulated in our population of COPD [70]. In turn, although not consistently shown, some authors have demonstrated an increase in local inflammation in the muscle of patients with COPD [71-73]. Regardless of the presence of inflammatory markers, activation of nuclear factor kB (NFkB) in peripheral muscle of patients with COPD suggests an increased inflammatory state [66,74-76]. TNFRS12A, a direct kB target, also known as TWEAK, is known to induce muscle wasting in whole muscle [77] and required for denervation atrophy [78], an effect mediated by NFkB [51,77]. TNFRS12A was up-regulated in COPDL and negatively correlated with FFMI and QMVC in our population.
The maintenance of skeletal muscle bulk results from the interaction of mechanisms leading muscle wasting (i.e. cell death, protein degradation) and muscle regeneration and protein synthesis. Several genes related to muscle development and regeneration (PMEPA1, IRX4, CHRDL2) were down-regulated in the COPDL [79,80]. Some groups have investigated key markers of muscle regeneration in peripheral muscle of patients with COPD, although the evidence is unclear due to the lack of longitudinal data. Plant et al. (80) showed no differences in skeletal muscle expression of muscle-specific transcription factors associated with muscle differentiation Myf5, MyoD or myogenin. Crul et al. [81] showed no differences in MyoD in stable COPD patients. However, patients undergoing an exacerbation presented with reduced levels of MyoD compared to healthy controls (78). Thériault et al. have recently shown that besides greater number of attempts to regenerate the muscle, there was a profound reduction in the differentiation potential in COPD patients peripheral muscle [82]. Vogiatzis et al. (123) showed that exercise training increased the expression of MyoD in peripheral muscle of patents with COPD. Lewis et al. (124) showed an increment in IGF-I protein (an activator of cell growth and proliferation and an inhibitor of apoptosis) with exercise training and a combination of exercise training and testosterone together with an increment in myogenin mRNA expression. The role of impaired muscle regeneration, potentially in some circumstances (e.g. exacerbations), remains to be elucidated.

Comparison with other studies

We have identified other studies assessing peripheral muscle gene expression in COPD and other medical conditions associated with muscle cachexia, namely intensive care (ICU) patients [83] and cancer [84], as well as ageing-associated muscle wasting [85,86] and studies exploring the effect of exercise training on gene expression in muscle of healthy subjects [87].
Despite differences in the populations and the methodology, which oblige to be cautious when comparing studies, we found interesting similarities with our results.
Two studies have explored gene expression in peripheral muscle of patients with COPD [88,89]. Debigaré [88] et al. compared vastus lateralis gene expression profiles from COPD patients with muscle atrophy and healthy controls. They have also found down regulation of genes involved in energy production and up-regulated genes related to cell cycle arrest including CDKN1A in the population of patients with COPD and muscle wasting. In contrast, while several relevant genes from our study were also identified by other studies involving stable COPD patients [88] this was not the case in studies involving hospitalized patients [89], however, as in our study, pro-inflammatory genes, such as SAA, were identified in the exacerbated patients.
In addition, our results have similarities with other inflammatory medical conditions associated with muscle wasting, such as sepsis [83], and also in association with ageing [85,86]. Similarly to our population, genes encoding proteins involved in muscle metabolism are down-regulated in association with ageing. Moreover, genes involved in cell cycle regulation and apoptosis are also affected by age. CDKN1A was up-regulated in the two mentioned studies in association with ageing. The similarities between our findings and studies exploring gene expression in ageing suggest that cell senescence may play a role as a pathogenic mechanism of muscle wasting in COPD. In fact, animal models of premature ageing show structural changes in the lungs and skeletal muscle that resemble those in COPD [90]. Shortening of telomeres, a marker of premature ageing, has been described in patients with COPD and muscle wasting [21].
Interestingly, as shown by Fredriksson et al. [83] in septic patients admitted to an intensive care unit (ICU), pro-inflammatory genes are up-regulated in the muscle of these patients including several genes found in our study. Moreover, they also found up-regulated genes related to cell cycle arrest including CDKN1A suggesting that that cell senescence could be also a mechanism leading to muscle wasting in this population. Moreover, inflammation and oxidative stress, a feature of patients with COPD and muscle wasting has been associated with stress-induced premature senescence [91].
Interestingly, when exploring the effects of exercise in skeletal muscle gene expression [86,87], while exercise training contribute to “normalise” the expression of genes related with energy production and oxidative capacity of the muscle, several genes found up-regulated in our COPD population were not modified (“normalized”) by exercise training (including GADD45A, NNMT, ANKRD1, ATF3 and SLC38A1) suggesting that the increased expression of these genes in our population of COPDL is not related to physical activity levels since this should produce the opposite effect.
It is of note that the gene profile of patients with cancer [84] was not comparable to the profile of patients with COPD and muscle wasting of our study proposing the possibility that muscle wasting in these two conditions involve different underlying mechanisms.

Limitations of the study

We have used FFMI measured by bioimpedance as a surrogate of muscle mass instead of a direct measurement of muscle mass. One of the most validated definitions of sarcopenia and severity of sarcopenia is based in BIA assessment of muscle mass [92]. This method has been validated against body composition assessed by deuterium dilution [93] and dual-energy X-ray absorptiometry [94] and shows very good correlation with other measurements of limb muscle mass in patients with COPD [95]. FFMI measured by bioimpedance is an independent predictor of skeletal muscle function and exercise capacity [96] and mortality [97] in patients with COPD. Moreover, it is a very sensitive method to detect undernutrition in these patients [98]. Values to differentiate muscle wasted patients from patients with preserved muscle mass based of FFMI have been established and validated in patients with COPD [93]. In our population, FFMI related to muscle function assessed as QMVC.
Physical activity (PA) can be seen as a confounding variable in this study. It was assessed with two questionnaires instead of using a direct measurement (i.e. activity monitors). PA assessed with the Voorrips questionnaire, designed to be use in elderly populations, showed lower physical activity levels in COPDL. It was difficult to recruit patients with higher levels of PA assessed with this tool. Interestingly, there was no difference in the level of Activity of Daily Living assessed with the London Chest Activity of Daily Living Scale. Moreover, key DEGs identified in the present study showed no modification in response to exercise training in young [87] and elderly [86] subjects making it less likely that these changes were attributed to a lower PA level in this population.
Beside the efforts in matching the populations, healthy control subjects present differences in average cessation days and Pack/years in comparison to COPDL. Environmental factors such as cigarette smoking may associate with peripheral muscle alterations [99]. However, it is unlikely to explain the differences in the transcriptome in our population as all three groups were matched for smoking status. Moreover, only one COPDL was an active smoker, whilst the other two groups included two active smokers each, which makes it less possible that this is a conditioning factor for muscle wasting in this group. Furthermore, no differences in the average cessation years, in cumulative history of smoking nor in age at smoking cessation (which may have important implications on outcomes) [100] were seen between COPDL and COPDN. Therefore, it is unlikely that the time post-smoking cessation contributes to the differences in gene expression between these groups.

Conclusions

This study demonstrate that vastus lateralis of patients with COPD and muscle wasting overexpress genes related to inhibition of cell cycle and of cell growth while genes related to muscle formation and growth and energy production were down-regulated. This pattern is similar to observations associated with ageing which, suggests that premature ageing may play a role in muscle atrophy in COPD. This profile, together with several genes involved in inflammation signaling, were shared with the profile described in severely ill patients in the ICU which suggest both, that ICU patients may also experience cell senescence in response to inflammation and that inflammation may be a shared mechanism between COPD and ICU patients. These results may open new avenues for the treatment of muscle wasting in patients with COPD. The most challenging issue is to explore potential avenues for treatment and identify the timing to treat these with anti-aging agents considering that earlier diagnosis is a key for effective anti-aging therapy.

Acknowledgments

The authors would like to thank the Chief Scientist Office (CSO 06/S1103/5) and the Fondo de Investigación Sanitaria (FIS 08/0320) for the financial support. The British Heart Foundation Centre of Research Excellence award is acknowledged for the data analysis support provided.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​4.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. 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.

Competing interests

The authors have no competing interests to declare. This study was financially supported by the Chief Scientist Office (CSO 06/S1103/5) and the Fondo de Investigación Sanitaria (FIS) (PI08/0320).

Authors’ contributions

Conception and design: RR, ED, RB, WM. Analysis and interpretation: RR, ED, JM, DD, MCDR, RB, WM. Drafting the manuscript for important intellectual contents: RR, ED, RB, WM. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Agusti AG, Noguera A, Sauleda J, Sala E, Pons J, Busquets X. Systemic effects of chronic obstructive pulmonary disease. Eur Respir J. 2003;21:347–60.CrossRefPubMed Agusti AG, Noguera A, Sauleda J, Sala E, Pons J, Busquets X. Systemic effects of chronic obstructive pulmonary disease. Eur Respir J. 2003;21:347–60.CrossRefPubMed
2.
Zurück zum Zitat Maltais F, Decramer M, Casaburi R, Barreiro E, Burelle Y, Debigare R, et al. An official american thoracic society/european respiratory society statement: update on limb muscle dysfunction in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2014;189:e15–62.CrossRefPubMedPubMedCentral Maltais F, Decramer M, Casaburi R, Barreiro E, Burelle Y, Debigare R, et al. An official american thoracic society/european respiratory society statement: update on limb muscle dysfunction in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2014;189:e15–62.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Decramer M, Gosselink R, Troosters T, Schepers R. Peripheral muscle weakness is associated with reduced survival in COPD. Am J Respir Crit Care Med. 1998;157:A19.CrossRef Decramer M, Gosselink R, Troosters T, Schepers R. Peripheral muscle weakness is associated with reduced survival in COPD. Am J Respir Crit Care Med. 1998;157:A19.CrossRef
4.
Zurück zum Zitat Engelen MPKJ, Schols AMWJ, Does JD, Wouters EFM. Skeletal muscle weakness is associated with wasting of extremity fat-free mass but not with airflow obstruction in patients with chronic obstructive pulmonary disease. Am J Clin Nutr. 2000;71:733–8.PubMed Engelen MPKJ, Schols AMWJ, Does JD, Wouters EFM. Skeletal muscle weakness is associated with wasting of extremity fat-free mass but not with airflow obstruction in patients with chronic obstructive pulmonary disease. Am J Clin Nutr. 2000;71:733–8.PubMed
5.
Zurück zum Zitat Bernard S, Leblanc P, Whittom F, Carrier G, Jobin J, Belleau R, et al. Peripheral muscle weakness in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;158:629–34.CrossRefPubMed Bernard S, Leblanc P, Whittom F, Carrier G, Jobin J, Belleau R, et al. Peripheral muscle weakness in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;158:629–34.CrossRefPubMed
6.
Zurück zum Zitat Vilaro J, Rabinovich R, Gonzalez-deSuso JM, Troosters T, Rodriguez D, Barbera JA, et al. Clinical assessment of peripheral muscle function in patients with chronic obstructive pulmonary disease. Am J Phys Med Rehabil. 2009;88:39–46.CrossRefPubMed Vilaro J, Rabinovich R, Gonzalez-deSuso JM, Troosters T, Rodriguez D, Barbera JA, et al. Clinical assessment of peripheral muscle function in patients with chronic obstructive pulmonary disease. Am J Phys Med Rehabil. 2009;88:39–46.CrossRefPubMed
7.
Zurück zum Zitat Schols AMWJ, Soeters PB, Dingemans AMC, Mostert R, Frantzen PJ, Wouters EFM. Prevalence and characteristics of nutritional depletion in patients with stable COPD eligible for pulmonary rehabilitation. Am Rev Respir Dis. 1993;147:1151–6.CrossRefPubMed Schols AMWJ, Soeters PB, Dingemans AMC, Mostert R, Frantzen PJ, Wouters EFM. Prevalence and characteristics of nutritional depletion in patients with stable COPD eligible for pulmonary rehabilitation. Am Rev Respir Dis. 1993;147:1151–6.CrossRefPubMed
8.
Zurück zum Zitat Baarends EM, Schols AM, Mostert R, Wouters EF. Peak exercise response in relation to tissue depletion in patients with chronic obstructive pulmonary disease. Eur Respir J. 1997;10:2807–13.CrossRefPubMed Baarends EM, Schols AM, Mostert R, Wouters EF. Peak exercise response in relation to tissue depletion in patients with chronic obstructive pulmonary disease. Eur Respir J. 1997;10:2807–13.CrossRefPubMed
9.
Zurück zum Zitat Kobayashi A, Yoneda T, Yoshikawa M, Ikuno M, Takenaka H, Fukuoka A, et al. The relation of fat-free mass to maximum exercise performance in patients with chronic obstructive pulmonary disease. Lung. 2000;178:119–27.CrossRefPubMed Kobayashi A, Yoneda T, Yoshikawa M, Ikuno M, Takenaka H, Fukuoka A, et al. The relation of fat-free mass to maximum exercise performance in patients with chronic obstructive pulmonary disease. Lung. 2000;178:119–27.CrossRefPubMed
10.
Zurück zum Zitat Schols AM, Mostert R, Soeters PB, Wouters EF. Body composition and exercise performance in patients with chronic obstructive pulmonary disease. Thorax. 1991;46:695–9.CrossRefPubMedPubMedCentral Schols AM, Mostert R, Soeters PB, Wouters EF. Body composition and exercise performance in patients with chronic obstructive pulmonary disease. Thorax. 1991;46:695–9.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Mostert R, Goris A, Weling-Scheepers C, Wouters EF, Schols AM. Tissue depletion and health related quality of life in patients with chronic obstructive pulmonary disease. Respir Med. 2000;94:859–67.CrossRefPubMed Mostert R, Goris A, Weling-Scheepers C, Wouters EF, Schols AM. Tissue depletion and health related quality of life in patients with chronic obstructive pulmonary disease. Respir Med. 2000;94:859–67.CrossRefPubMed
12.
Zurück zum Zitat Marquis K, Debigare R, Lacasse Y, Leblanc P, Jobin J, Carrier G, et al. Midthigh muscle cross-sectional area is a better predictor of mortality than body mass index in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2002;166:809–13.CrossRefPubMed Marquis K, Debigare R, Lacasse Y, Leblanc P, Jobin J, Carrier G, et al. Midthigh muscle cross-sectional area is a better predictor of mortality than body mass index in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2002;166:809–13.CrossRefPubMed
13.
Zurück zum Zitat Mador MJ. Muscle mass, not body weight, predicts outcome in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2002;166:787–9.CrossRefPubMed Mador MJ. Muscle mass, not body weight, predicts outcome in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2002;166:787–9.CrossRefPubMed
14.
Zurück zum Zitat Engelen MPKJ, Schols AMWJ, Baken WC, Wesseling GJ, Wouters EFM. Nutritional depletion in relation to respiratory and peripheral skeletal muscle function in out-patients with COPD. Eur Respir J. 1994;7:1793–7.CrossRefPubMed Engelen MPKJ, Schols AMWJ, Baken WC, Wesseling GJ, Wouters EFM. Nutritional depletion in relation to respiratory and peripheral skeletal muscle function in out-patients with COPD. Eur Respir J. 1994;7:1793–7.CrossRefPubMed
15.
Zurück zum Zitat Eid AA, Ionescu AA, Nixon LS, Lewis-Jenkins V, Matthews SB, Griffiths TL, et al. Inflammatory response and body composition in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2001;164:1414–8.CrossRefPubMed Eid AA, Ionescu AA, Nixon LS, Lewis-Jenkins V, Matthews SB, Griffiths TL, et al. Inflammatory response and body composition in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2001;164:1414–8.CrossRefPubMed
16.
Zurück zum Zitat Jakobsson P, Jorfeldt L, Brundin A. Skeletal muscle metabolits and fibre types in patients with advanced chronic obstructive pulmonary disease (COPD), with and without chronic respiratory failure. Eur Respir J. 1990;3:192–6.PubMed Jakobsson P, Jorfeldt L, Brundin A. Skeletal muscle metabolits and fibre types in patients with advanced chronic obstructive pulmonary disease (COPD), with and without chronic respiratory failure. Eur Respir J. 1990;3:192–6.PubMed
17.
Zurück zum Zitat Simard C, Maltais F, Leblanc P, Simard P, Jobin J. Mitochondrial and capillarity changes in vastus lateralis muscle of COPD patients: electron microscopy study. Med Sci Sports Exerc. 1996;28:S95.CrossRef Simard C, Maltais F, Leblanc P, Simard P, Jobin J. Mitochondrial and capillarity changes in vastus lateralis muscle of COPD patients: electron microscopy study. Med Sci Sports Exerc. 1996;28:S95.CrossRef
18.
Zurück zum Zitat Rabinovich RA, Bastos R, Ardite E, Orozco-Levi M, Gea J, Vilar¢ J, et al. Mitochondrial dysfunction in COPD patients with low body mass index. Eur Respir J. 2007;29:643–50.CrossRefPubMed Rabinovich RA, Bastos R, Ardite E, Orozco-Levi M, Gea J, Vilar¢ J, et al. Mitochondrial dysfunction in COPD patients with low body mass index. Eur Respir J. 2007;29:643–50.CrossRefPubMed
19.
Zurück zum Zitat Puente-Maestu L, Perez-Parra J, Godoy R, Moreno N, Tejedor A, Gonzalez-Aragoneses F, et al. Abnormal mitochondrial function in locomotor and respiratory muscles of COPD patients. Eur Respir J. 2009;33:1045–52.CrossRefPubMed Puente-Maestu L, Perez-Parra J, Godoy R, Moreno N, Tejedor A, Gonzalez-Aragoneses F, et al. Abnormal mitochondrial function in locomotor and respiratory muscles of COPD patients. Eur Respir J. 2009;33:1045–52.CrossRefPubMed
21.
22.
Zurück zum Zitat Bernard S, Whittom F, Leblanc P, Jobin J, Belleau R, Berube C, et al. Aerobic and strength training in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1999;159:896–901.CrossRefPubMed Bernard S, Whittom F, Leblanc P, Jobin J, Belleau R, Berube C, et al. Aerobic and strength training in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1999;159:896–901.CrossRefPubMed
23.
Zurück zum Zitat Decramer M. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease (updated 2014). ᅟ. 2014;ᅟ:ᅟ. www.goldcopd.com. Decramer M. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease (updated 2014). ᅟ. 2014;ᅟ:ᅟ. www.​goldcopd.​com.
24.
Zurück zum Zitat ATS Committee on Proficiency Standards for Clinical Pulmonary Function Laboratories. ATS statement: guidelines for the six-minute walk test. Am J Respir Crit Care Med. 2002;166:111–7.CrossRef ATS Committee on Proficiency Standards for Clinical Pulmonary Function Laboratories. ATS statement: guidelines for the six-minute walk test. Am J Respir Crit Care Med. 2002;166:111–7.CrossRef
25.
Zurück zum Zitat Spruit MA, Gosselink R, Troosters T, Kasran A, Gayan-Ramirez G, Bogaerts P, et al. Muscle force during an acute exacerbation in hospitalised patients with COPD and its relationship with CXCL8 and IGF-I. Thorax. 2003;58:752–6.CrossRefPubMedPubMedCentral Spruit MA, Gosselink R, Troosters T, Kasran A, Gayan-Ramirez G, Bogaerts P, et al. Muscle force during an acute exacerbation in hospitalised patients with COPD and its relationship with CXCL8 and IGF-I. Thorax. 2003;58:752–6.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Jones PW, Quirk FH, Baveystock CM, Littlejohns P. A self-complete measure of health status for chronic airflow limitation. Am Rev Respir Dis. 1992;145:1321–7.CrossRefPubMed Jones PW, Quirk FH, Baveystock CM, Littlejohns P. A self-complete measure of health status for chronic airflow limitation. Am Rev Respir Dis. 1992;145:1321–7.CrossRefPubMed
27.
Zurück zum Zitat Voorrips LE, Ravelli AC, Dongelmans PC, Deurenberg P, Van Staveren WA. A physical activity questionnaire for the elderly. Med Sci Sports Exerc. 1991;23:974–9.CrossRefPubMed Voorrips LE, Ravelli AC, Dongelmans PC, Deurenberg P, Van Staveren WA. A physical activity questionnaire for the elderly. Med Sci Sports Exerc. 1991;23:974–9.CrossRefPubMed
28.
Zurück zum Zitat Garrod R, Bestall JC, Paul EA, Wedzicha JA, Jones PW. Development and validation of a standardized measure of activity of daily living in patients with severe COPD: the London chest activity of daily living scale (LCADL). Respir Med. 2000;94:589–96.CrossRefPubMed Garrod R, Bestall JC, Paul EA, Wedzicha JA, Jones PW. Development and validation of a standardized measure of activity of daily living in patients with severe COPD: the London chest activity of daily living scale (LCADL). Respir Med. 2000;94:589–96.CrossRefPubMed
29.
Zurück zum Zitat Schols AM, Broekhuizen R, Weling-Scheepers CA, Wouters EF. Body composition and mortality in chronic obstructive pulmonary disease. Am J Clin Nutr. 2005;82:53–9.PubMed Schols AM, Broekhuizen R, Weling-Scheepers CA, Wouters EF. Body composition and mortality in chronic obstructive pulmonary disease. Am J Clin Nutr. 2005;82:53–9.PubMed
30.
Zurück zum Zitat Toth ZE, Mezey E. Simultaneous visualization of multiple antigens with tyramide signal amplification using antibodies from the same species. J Histochem Cytochem. 2007;55:545–54.CrossRefPubMed Toth ZE, Mezey E. Simultaneous visualization of multiple antigens with tyramide signal amplification using antibodies from the same species. J Histochem Cytochem. 2007;55:545–54.CrossRefPubMed
31.
Zurück zum Zitat Team RDC. A Language and Environment for Statistical Computing. R Foundation for Statistical Computing. In: Book A Language and Environment for Statistical Computing. Vienna, Austria: R Foundation for Statistical Computing; 2005. Team RDC. A Language and Environment for Statistical Computing. R Foundation for Statistical Computing. In: Book A Language and Environment for Statistical Computing. Vienna, Austria: R Foundation for Statistical Computing; 2005.
32.
Zurück zum Zitat Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 2004;5:R80.CrossRefPubMedPubMedCentral Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 2004;5:R80.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Breitling R, Armengaud P, Amtmann A, Herzyk P. Rank products: a simple, yet powerful, new method to detect differentially regulated genes in replicated microarray experiments. FEBS Lett. 2004;573:83–92.CrossRefPubMed Breitling R, Armengaud P, Amtmann A, Herzyk P. Rank products: a simple, yet powerful, new method to detect differentially regulated genes in replicated microarray experiments. FEBS Lett. 2004;573:83–92.CrossRefPubMed
34.
Zurück zum Zitat Hong F, Breitling R, McEntee CW, Wittner BS, Nemhauser JL, Chory J. RankProd: a bioconductor package for detecting differentially expressed genes in meta-analysis. Bioinformatics. 2006;22:2825–7.CrossRefPubMed Hong F, Breitling R, McEntee CW, Wittner BS, Nemhauser JL, Chory J. RankProd: a bioconductor package for detecting differentially expressed genes in meta-analysis. Bioinformatics. 2006;22:2825–7.CrossRefPubMed
35.
Zurück zum Zitat Gallagher IJ, Scheele C, Keller P, Nielsen AR, Remenyi J, Fischer CP, et al. Integration of microrna changes in vivo identifies novel molecular features of muscle insulin resistance in type 2 diabetes. Genome Med. 2010;2:9.CrossRefPubMedPubMedCentral Gallagher IJ, Scheele C, Keller P, Nielsen AR, Remenyi J, Fischer CP, et al. Integration of microrna changes in vivo identifies novel molecular features of muscle insulin resistance in type 2 diabetes. Genome Med. 2010;2:9.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Calvano SE, Xiao W, Richards DR, Felciano RM, Baker HV, Cho RJ, et al. A network-based analysis of systemic inflammation in humans. Nature. 2005;437:1032–7.CrossRefPubMed Calvano SE, Xiao W, Richards DR, Felciano RM, Baker HV, Cho RJ, et al. A network-based analysis of systemic inflammation in humans. Nature. 2005;437:1032–7.CrossRefPubMed
37.
Zurück zum Zitat Doucet M, Russell AP, Leger B, Debigare R, Joanisse DR, Caron MA, et al. Muscle atrophy and hypertrophy signaling in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2007;176:261–9.CrossRefPubMed Doucet M, Russell AP, Leger B, Debigare R, Joanisse DR, Caron MA, et al. Muscle atrophy and hypertrophy signaling in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2007;176:261–9.CrossRefPubMed
38.
Zurück zum Zitat Dong C, Li Q, Lyu SC, Krensky AM, Clayberger C. A novel apoptosis pathway activated by the carboxyl terminus of p21. Blood. 2005;105:1187–94.CrossRefPubMed Dong C, Li Q, Lyu SC, Krensky AM, Clayberger C. A novel apoptosis pathway activated by the carboxyl terminus of p21. Blood. 2005;105:1187–94.CrossRefPubMed
39.
Zurück zum Zitat Ebert SM, Dyle MC, Kunkel SD, Bullard SA, Bongers KS, Fox DK, et al. Stress-induced skeletal muscle Gadd45a expression reprograms myonuclei and causes muscle atrophy. J Biol Chem. 2012;287:27290–301.CrossRefPubMedPubMedCentral Ebert SM, Dyle MC, Kunkel SD, Bullard SA, Bongers KS, Fox DK, et al. Stress-induced skeletal muscle Gadd45a expression reprograms myonuclei and causes muscle atrophy. J Biol Chem. 2012;287:27290–301.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Caiozzo VJ, Utkan A, Chou R, Khalafi A, Chandra H, Baker M, et al. Effects of distraction on muscle length: mechanisms involved in sarcomerogenesis. Clin Orthop Relat Res 2002:S133-145 Caiozzo VJ, Utkan A, Chou R, Khalafi A, Chandra H, Baker M, et al. Effects of distraction on muscle length: mechanisms involved in sarcomerogenesis. Clin Orthop Relat Res 2002:S133-145
41.
Zurück zum Zitat Bongers KS, Fox DK, Ebert SM, Kunkel SD, Dyle MC, Bullard SA, et al. Skeletal muscle denervation causes skeletal muscle atrophy through a pathway that involves both Gadd45a and HDAC4. Am J Physiol Endocrinol Metab. 2013;305:E907–15.CrossRefPubMedPubMedCentral Bongers KS, Fox DK, Ebert SM, Kunkel SD, Dyle MC, Bullard SA, et al. Skeletal muscle denervation causes skeletal muscle atrophy through a pathway that involves both Gadd45a and HDAC4. Am J Physiol Endocrinol Metab. 2013;305:E907–15.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Calura E, Cagnin S, Raffaello A, Laveder P, Lanfranchi G, Romualdi C. Meta-analysis of expression signatures of muscle atrophy: gene interaction networks in early and late stages. BMC Genomics. 2008;9:630.CrossRefPubMedPubMedCentral Calura E, Cagnin S, Raffaello A, Laveder P, Lanfranchi G, Romualdi C. Meta-analysis of expression signatures of muscle atrophy: gene interaction networks in early and late stages. BMC Genomics. 2008;9:630.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat De Aguilar JL G, Niederhauser-Wiederkehr C, Halter B, De Tapia M, Di Scala F, Demougin P, et al. Gene profiling of skeletal muscle in an amyotrophic lateral sclerosis mouse model. Physiol Genomics. 2008;32:207–18.CrossRef De Aguilar JL G, Niederhauser-Wiederkehr C, Halter B, De Tapia M, Di Scala F, Demougin P, et al. Gene profiling of skeletal muscle in an amyotrophic lateral sclerosis mouse model. Physiol Genomics. 2008;32:207–18.CrossRef
44.
Zurück zum Zitat Ohsawa Y, Hagiwara H, Nakatani M, Yasue A, Moriyama K, Murakami T, et al. Muscular atrophy of caveolin-3-deficient mice is rescued by myostatin inhibition. J Clin Invest. 2006;116:2924–34.CrossRefPubMedPubMedCentral Ohsawa Y, Hagiwara H, Nakatani M, Yasue A, Moriyama K, Murakami T, et al. Muscular atrophy of caveolin-3-deficient mice is rescued by myostatin inhibition. J Clin Invest. 2006;116:2924–34.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Thomas M, Langley B, Berry C, Sharma M, Kirk S, Bass J, et al. Myostatin, a negative regulator of muscle growth, functions by inhibiting myoblast proliferation. J Biol Chem. 2000;275:40235–43.CrossRefPubMed Thomas M, Langley B, Berry C, Sharma M, Kirk S, Bass J, et al. Myostatin, a negative regulator of muscle growth, functions by inhibiting myoblast proliferation. J Biol Chem. 2000;275:40235–43.CrossRefPubMed
46.
Zurück zum Zitat Dotto GP. p21(WAF1/Cip1): more than a break to the cell cycle? Biochim Biophys Acta. 2000;1471:M43–56.PubMed Dotto GP. p21(WAF1/Cip1): more than a break to the cell cycle? Biochim Biophys Acta. 2000;1471:M43–56.PubMed
47.
Zurück zum Zitat Gartel AL, Tyner AL. The role of the cyclin-dependent kinase inhibitor p21 in apoptosis. Mol Cancer Ther. 2002;1:639–49.PubMed Gartel AL, Tyner AL. The role of the cyclin-dependent kinase inhibitor p21 in apoptosis. Mol Cancer Ther. 2002;1:639–49.PubMed
48.
Zurück zum Zitat Wang A, Gu J, Judson-Kremer K, Powell KL, Mistry H, Simhambhatla P, et al. Response of human mammary epithelial cells to DNA damage induced by BPDE: involvement of novel regulatory pathways. Carcinogenesis. 2003;24:225–34.CrossRefPubMed Wang A, Gu J, Judson-Kremer K, Powell KL, Mistry H, Simhambhatla P, et al. Response of human mammary epithelial cells to DNA damage induced by BPDE: involvement of novel regulatory pathways. Carcinogenesis. 2003;24:225–34.CrossRefPubMed
49.
Zurück zum Zitat Okamoto Y, Chaves A, Chen J, Kelley R, Jones K, Weed HG, et al. Transgenic mice with cardiac-specific expression of activating transcription factor 3, a stress-inducible gene, have conduction abnormalities and contractile dysfunction. Am J Pathol. 2001;159:639–50.CrossRefPubMedPubMedCentral Okamoto Y, Chaves A, Chen J, Kelley R, Jones K, Weed HG, et al. Transgenic mice with cardiac-specific expression of activating transcription factor 3, a stress-inducible gene, have conduction abnormalities and contractile dysfunction. Am J Pathol. 2001;159:639–50.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Laure L, Suel L, Roudaut C, Bourg N, Ouali A, Bartoli M, et al. Cardiac ankyrin repeat protein is a marker of skeletal muscle pathological remodelling. FEBS J. 2009;276:669–84.CrossRefPubMed Laure L, Suel L, Roudaut C, Bourg N, Ouali A, Bartoli M, et al. Cardiac ankyrin repeat protein is a marker of skeletal muscle pathological remodelling. FEBS J. 2009;276:669–84.CrossRefPubMed
51.
Zurück zum Zitat Wu CL, Kandarian SC, Jackman RW. Identification of genes that elicit disuse muscle atrophy via the transcription factors p50 and Bcl-3. PloS One. 2011;6:e16171.CrossRefPubMedPubMedCentral Wu CL, Kandarian SC, Jackman RW. Identification of genes that elicit disuse muscle atrophy via the transcription factors p50 and Bcl-3. PloS One. 2011;6:e16171.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Timchenko NA, Wilde M, Nakanishi M, Smith JR, Darlington GJ. CCAAT/enhancer-binding protein alpha (C/EBP alpha) inhibits cell proliferation through the p21 (WAF-1/CIP-1/SDI-1) protein. Genes Dev. 1996;10:804–15.CrossRefPubMed Timchenko NA, Wilde M, Nakanishi M, Smith JR, Darlington GJ. CCAAT/enhancer-binding protein alpha (C/EBP alpha) inhibits cell proliferation through the p21 (WAF-1/CIP-1/SDI-1) protein. Genes Dev. 1996;10:804–15.CrossRefPubMed
53.
Zurück zum Zitat Timchenko NA, Harris TE, Wilde M, Bilyeu TA, Burgess-Beusse BL, Finegold MJ, et al. CCAAT/enhancer binding protein alpha regulates p21 protein and hepatocyte proliferation in newborn mice. Mol Cell Biol. 1997;17:7353–61.CrossRefPubMedPubMedCentral Timchenko NA, Harris TE, Wilde M, Bilyeu TA, Burgess-Beusse BL, Finegold MJ, et al. CCAAT/enhancer binding protein alpha regulates p21 protein and hepatocyte proliferation in newborn mice. Mol Cell Biol. 1997;17:7353–61.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Agusti AG, Sauleda J, Miralles C, Gomez C, Togores B, Sala E, et al. Skeletal muscle apoptosis and weight loss in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2002;166:485–9.CrossRefPubMed Agusti AG, Sauleda J, Miralles C, Gomez C, Togores B, Sala E, et al. Skeletal muscle apoptosis and weight loss in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2002;166:485–9.CrossRefPubMed
55.
Zurück zum Zitat Barreiro E, Ferrer D, Sanchez F, Minguella J, Marin-Corral J, Martinez-Llorens J, et al. Inflammatory cells and apoptosis in respiratory and limb muscles of patients with COPD. J Appl Physiol. 2011;111:808–17.CrossRefPubMed Barreiro E, Ferrer D, Sanchez F, Minguella J, Marin-Corral J, Martinez-Llorens J, et al. Inflammatory cells and apoptosis in respiratory and limb muscles of patients with COPD. J Appl Physiol. 2011;111:808–17.CrossRefPubMed
56.
Zurück zum Zitat Gosker HR, Kubat B, Schaart G, van der Vusse GJ, Wouters EF, Schols AM. Myopathological features in skeletal muscle of patients with chronic obstructive pulmonary disease. Eur Respir J. 2003;22:280–5.CrossRefPubMed Gosker HR, Kubat B, Schaart G, van der Vusse GJ, Wouters EF, Schols AM. Myopathological features in skeletal muscle of patients with chronic obstructive pulmonary disease. Eur Respir J. 2003;22:280–5.CrossRefPubMed
57.
Zurück zum Zitat Suzuki A, Tsutomi Y, Akahane K, Araki T, Miura M. Resistance to Fas-mediated apoptosis: activation of caspase 3 is regulated by cell cycle regulator p21WAF1 and IAP gene family ILP. Oncogene. 1998;17:931–9.CrossRefPubMed Suzuki A, Tsutomi Y, Akahane K, Araki T, Miura M. Resistance to Fas-mediated apoptosis: activation of caspase 3 is regulated by cell cycle regulator p21WAF1 and IAP gene family ILP. Oncogene. 1998;17:931–9.CrossRefPubMed
58.
Zurück zum Zitat Hubal MJ, Chen TC, Thompson PD, Clarkson PM. Inflammatory gene changes associated with the repeated-bout effect. Am J Physiol Regul Integr Comp Physiol. 2008;294:R1628–37.CrossRefPubMed Hubal MJ, Chen TC, Thompson PD, Clarkson PM. Inflammatory gene changes associated with the repeated-bout effect. Am J Physiol Regul Integr Comp Physiol. 2008;294:R1628–37.CrossRefPubMed
59.
Zurück zum Zitat Kivela R, Kyrolainen H, Selanne H, Komi PV, Kainulainen H, Vihko V. A single bout of exercise with high mechanical loading induces the expression of Cyr61/CCN1 and CTGF/CCN2 in human skeletal muscle. J Appl Physiol (1985). 2007;103:1395–401.CrossRef Kivela R, Kyrolainen H, Selanne H, Komi PV, Kainulainen H, Vihko V. A single bout of exercise with high mechanical loading induces the expression of Cyr61/CCN1 and CTGF/CCN2 in human skeletal muscle. J Appl Physiol (1985). 2007;103:1395–401.CrossRef
60.
Zurück zum Zitat Kivela R, Silvennoinen M, Lehti M, Jalava S, Vihko V, Kainulainen H. Exercise-induced expression of angiogenic growth factors in skeletal muscle and in capillaries of healthy and diabetic mice. Cardiovasc Diabetol. 2008;7:13.CrossRefPubMedPubMedCentral Kivela R, Silvennoinen M, Lehti M, Jalava S, Vihko V, Kainulainen H. Exercise-induced expression of angiogenic growth factors in skeletal muscle and in capillaries of healthy and diabetic mice. Cardiovasc Diabetol. 2008;7:13.CrossRefPubMedPubMedCentral
61.
Zurück zum Zitat O’Connor RS, Mills ST, Jones KA, Ho SN, Pavlath GK. A combinatorial role for NFAT5 in both myoblast migration and differentiation during skeletal muscle myogenesis. J Cell Sci. 2007;120:149–59.CrossRefPubMed O’Connor RS, Mills ST, Jones KA, Ho SN, Pavlath GK. A combinatorial role for NFAT5 in both myoblast migration and differentiation during skeletal muscle myogenesis. J Cell Sci. 2007;120:149–59.CrossRefPubMed
62.
Zurück zum Zitat Rayssac A, Neveu C, Pucelle M, Van den Berghe L, Prado-Lourenco L, Arnal JF, et al. IRES-based vector coexpressing FGF2 and Cyr61 provides synergistic and safe therapeutics of lower limb ischemia. Mol Ther. 2009;17:2010–9.CrossRefPubMedPubMedCentral Rayssac A, Neveu C, Pucelle M, Van den Berghe L, Prado-Lourenco L, Arnal JF, et al. IRES-based vector coexpressing FGF2 and Cyr61 provides synergistic and safe therapeutics of lower limb ischemia. Mol Ther. 2009;17:2010–9.CrossRefPubMedPubMedCentral
63.
Zurück zum Zitat Lehnert SA, Byrne KA, Reverter A, Nattrass GS, Greenwood PL, Wang YH, et al. Gene expression profiling of bovine skeletal muscle in response to and during recovery from chronic and severe undernutrition. J Anim Sci. 2006;84:3239–50.CrossRefPubMed Lehnert SA, Byrne KA, Reverter A, Nattrass GS, Greenwood PL, Wang YH, et al. Gene expression profiling of bovine skeletal muscle in response to and during recovery from chronic and severe undernutrition. J Anim Sci. 2006;84:3239–50.CrossRefPubMed
64.
Zurück zum Zitat Magnusson C, Svensson A, Christerson U, Tagerud S. Denervation-induced alterations in gene expression in mouse skeletal muscle. Eur J Neurosci. 2005;21:577–80.CrossRefPubMed Magnusson C, Svensson A, Christerson U, Tagerud S. Denervation-induced alterations in gene expression in mouse skeletal muscle. Eur J Neurosci. 2005;21:577–80.CrossRefPubMed
65.
Zurück zum Zitat Lobel M, Bauer S, Meisel C, Eisenreich A, Kudernatsch R, Tank J, et al. Ccn1: a novel inflammation-regulated biphasic immune cell migration modulator. Cellular and molecular life sciences : CMLS. 2012;69:3101–13.CrossRefPubMed Lobel M, Bauer S, Meisel C, Eisenreich A, Kudernatsch R, Tank J, et al. Ccn1: a novel inflammation-regulated biphasic immune cell migration modulator. Cellular and molecular life sciences : CMLS. 2012;69:3101–13.CrossRefPubMed
66.
Zurück zum Zitat Fermoselle C, Rabinovich R, Ausin P, Puig-Vilanova E, Coronell C, Sanchez F, et al. Does oxidative stress modulate limb muscle atrophy in severe COPD patients? Eur Respir J. 2012;40:851–62.CrossRefPubMed Fermoselle C, Rabinovich R, Ausin P, Puig-Vilanova E, Coronell C, Sanchez F, et al. Does oxidative stress modulate limb muscle atrophy in severe COPD patients? Eur Respir J. 2012;40:851–62.CrossRefPubMed
67.
Zurück zum Zitat Barreiro E, Rabinovich R, Marin-Corral J, Barbera JA, Gea J, Roca J. Chronic endurance exercise induces quadriceps nitrosative stress in patients with severe COPD. Thorax. 2009;64:13–9.CrossRefPubMed Barreiro E, Rabinovich R, Marin-Corral J, Barbera JA, Gea J, Roca J. Chronic endurance exercise induces quadriceps nitrosative stress in patients with severe COPD. Thorax. 2009;64:13–9.CrossRefPubMed
68.
Zurück zum Zitat Rabinovich RA, Ardite E, Troosters T, Carb¢ N, Alonso J, de Suso JM G, et al. Reduced muscle redox capacity after endurance training in copd patients. American Journal of Respiratory and Critical Care Medicine. 2001;164:1114–8.CrossRefPubMed Rabinovich RA, Ardite E, Troosters T, Carb¢ N, Alonso J, de Suso JM G, et al. Reduced muscle redox capacity after endurance training in copd patients. American Journal of Respiratory and Critical Care Medicine. 2001;164:1114–8.CrossRefPubMed
69.
Zurück zum Zitat Rabinovich RA, Ardite E, Mayer AM, Figueras Polo M, Vilar¢ J, Argiles JM, et al. Training depletes muscle glutathione in COPD patients with low body mass index. Respiration. 2006;73:757–61.CrossRefPubMed Rabinovich RA, Ardite E, Mayer AM, Figueras Polo M, Vilar¢ J, Argiles JM, et al. Training depletes muscle glutathione in COPD patients with low body mass index. Respiration. 2006;73:757–61.CrossRefPubMed
70.
Zurück zum Zitat Paine A, Eiz-Vesper B, Blasczyk R, Immenschuh S. Signaling to heme oxygenase-1 and its anti-inflammatory therapeutic potential. Biochem Pharmacol. 2010;80:1895–903.CrossRefPubMed Paine A, Eiz-Vesper B, Blasczyk R, Immenschuh S. Signaling to heme oxygenase-1 and its anti-inflammatory therapeutic potential. Biochem Pharmacol. 2010;80:1895–903.CrossRefPubMed
71.
Zurück zum Zitat Rabinovich RA, Figueras M, Ardite E, Carbo N, Troosters T, Filella X, et al. Increased tumour necrosis factor-alpha plasma levels during moderate-intensity exercise in COPD patients. Eur Respir J. 2003;21:789–94.CrossRefPubMed Rabinovich RA, Figueras M, Ardite E, Carbo N, Troosters T, Filella X, et al. Increased tumour necrosis factor-alpha plasma levels during moderate-intensity exercise in COPD patients. Eur Respir J. 2003;21:789–94.CrossRefPubMed
72.
Zurück zum Zitat Montes De Oca M, Torres SH, De Sanctis J, Mata A, Hernandez N, Talamo C. Skeletal muscle inflammation and nitric oxide in patients with COPD. Eur Respir J. 2005;26:390–7.CrossRefPubMed Montes De Oca M, Torres SH, De Sanctis J, Mata A, Hernandez N, Talamo C. Skeletal muscle inflammation and nitric oxide in patients with COPD. Eur Respir J. 2005;26:390–7.CrossRefPubMed
73.
Zurück zum Zitat Remels AH, Gosker HR, Schrauwen P, Hommelberg PP, Sliwinski P, Polkey M, et al. TNF-alpha impairs regulation of muscle oxidative phenotype: implications for cachexia? FASEB J. 2010;24:5052–62.CrossRefPubMed Remels AH, Gosker HR, Schrauwen P, Hommelberg PP, Sliwinski P, Polkey M, et al. TNF-alpha impairs regulation of muscle oxidative phenotype: implications for cachexia? FASEB J. 2010;24:5052–62.CrossRefPubMed
74.
Zurück zum Zitat Agusti A, Morla M, Sauleda J, Saus C, Busquets X. NF-kappaB activation and iNOS upregulation in skeletal muscle of patients with COPD and low body weight. Thorax. 2004;59:483–7.CrossRefPubMedPubMedCentral Agusti A, Morla M, Sauleda J, Saus C, Busquets X. NF-kappaB activation and iNOS upregulation in skeletal muscle of patients with COPD and low body weight. Thorax. 2004;59:483–7.CrossRefPubMedPubMedCentral
75.
Zurück zum Zitat Plant PJ, Brooks D, Faughnan M, Bayley T, Bain J, Singer L, et al. Cellular markers of muscle atrophy in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2010;42:461–71.CrossRefPubMed Plant PJ, Brooks D, Faughnan M, Bayley T, Bain J, Singer L, et al. Cellular markers of muscle atrophy in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2010;42:461–71.CrossRefPubMed
76.
Zurück zum Zitat Vogiatzis I, Simoes DC, Stratakos G, Kourepini E, Terzis G, Manta P, et al. Effect of pulmonary rehabilitation on muscle remodelling in cachectic patients with COPD. Eur Respir J. 2010;36:301–10.CrossRefPubMed Vogiatzis I, Simoes DC, Stratakos G, Kourepini E, Terzis G, Manta P, et al. Effect of pulmonary rehabilitation on muscle remodelling in cachectic patients with COPD. Eur Respir J. 2010;36:301–10.CrossRefPubMed
77.
Zurück zum Zitat Dogra C, Changotra H, Wedhas N, Qin X, Wergedal JE, Kumar A. TNF-related weak inducer of apoptosis (TWEAK) is a potent skeletal muscle-wasting cytokine. FASEB J. 2007;21:1857–69.CrossRefPubMedPubMedCentral Dogra C, Changotra H, Wedhas N, Qin X, Wergedal JE, Kumar A. TNF-related weak inducer of apoptosis (TWEAK) is a potent skeletal muscle-wasting cytokine. FASEB J. 2007;21:1857–69.CrossRefPubMedPubMedCentral
78.
Zurück zum Zitat Mittal A, Bhatnagar S, Kumar A, Lach-Trifilieff E, Wauters S, Li H, et al. The TWEAK-Fn14 system is a critical regulator of denervation-induced skeletal muscle atrophy in mice. J Cell Biol. 2010;188:833–49.CrossRefPubMedPubMedCentral Mittal A, Bhatnagar S, Kumar A, Lach-Trifilieff E, Wauters S, Li H, et al. The TWEAK-Fn14 system is a critical regulator of denervation-induced skeletal muscle atrophy in mice. J Cell Biol. 2010;188:833–49.CrossRefPubMedPubMedCentral
79.
Zurück zum Zitat Park CY, Pierce SA, Von Drehle M, Ivey KN, Morgan JA, Blau HM, et al. skNAC, a Smyd1-interacting transcription factor, is involved in cardiac development and skeletal muscle growth and regeneration. Proc Natl Acad Sci U S A. 2010;107:20750–5.CrossRefPubMedPubMedCentral Park CY, Pierce SA, Von Drehle M, Ivey KN, Morgan JA, Blau HM, et al. skNAC, a Smyd1-interacting transcription factor, is involved in cardiac development and skeletal muscle growth and regeneration. Proc Natl Acad Sci U S A. 2010;107:20750–5.CrossRefPubMedPubMedCentral
80.
Zurück zum Zitat Wu Z, Nagano I, Takahashi Y. Candidate genes responsible for common and different pathology of infected muscle tissues between Trichinella spiralis and T. pseudospiralis infection. Parasitol Int. 2008;57:368–78.CrossRefPubMed Wu Z, Nagano I, Takahashi Y. Candidate genes responsible for common and different pathology of infected muscle tissues between Trichinella spiralis and T. pseudospiralis infection. Parasitol Int. 2008;57:368–78.CrossRefPubMed
81.
Zurück zum Zitat Crul T, Spruit MA, Gayan-Ramirez G, Quarck R, Gosselink R, Troosters T, et al. Markers of inflammation and disuse in vastus lateralis of chronic obstructive pulmonary disease patients. Eur J Clin Invest. 2007;37:897–904.CrossRefPubMed Crul T, Spruit MA, Gayan-Ramirez G, Quarck R, Gosselink R, Troosters T, et al. Markers of inflammation and disuse in vastus lateralis of chronic obstructive pulmonary disease patients. Eur J Clin Invest. 2007;37:897–904.CrossRefPubMed
82.
Zurück zum Zitat Theriault ME, Pare ME, Lemire BB, Maltais F, Debigare R. Regenerative defect in vastus lateralis muscle of patients with chronic obstructive pulmonary disease. Respir Res. 2014;15:35.CrossRefPubMedPubMedCentral Theriault ME, Pare ME, Lemire BB, Maltais F, Debigare R. Regenerative defect in vastus lateralis muscle of patients with chronic obstructive pulmonary disease. Respir Res. 2014;15:35.CrossRefPubMedPubMedCentral
83.
Zurück zum Zitat Fredriksson K, Tjader I, Keller P, Petrovic N, Ahlman B, Scheele C, et al. Dysregulation of mitochondrial dynamics and the muscle transcriptome in ICU patients suffering from sepsis induced multiple organ failure. PloS One. 2008;3:e3686.CrossRefPubMedPubMedCentral Fredriksson K, Tjader I, Keller P, Petrovic N, Ahlman B, Scheele C, et al. Dysregulation of mitochondrial dynamics and the muscle transcriptome in ICU patients suffering from sepsis induced multiple organ failure. PloS One. 2008;3:e3686.CrossRefPubMedPubMedCentral
84.
Zurück zum Zitat Stephens NA, Gallagher IJ, Rooyackers O, Skipworth RJ, Tan BH, Marstrand T, et al. Using transcriptomics to identify and validate novel biomarkers of human skeletal muscle cancer cachexia. Genome Med. 2010;2:1.CrossRefPubMedPubMedCentral Stephens NA, Gallagher IJ, Rooyackers O, Skipworth RJ, Tan BH, Marstrand T, et al. Using transcriptomics to identify and validate novel biomarkers of human skeletal muscle cancer cachexia. Genome Med. 2010;2:1.CrossRefPubMedPubMedCentral
85.
Zurück zum Zitat Welle S, Brooks AI, Delehanty JM, Needler N, Bhatt K, Shah B, et al. Skeletal muscle gene expression profiles in 20–29 year old and 65–71 year old women. Exp Gerontol. 2004;39:369–77.CrossRefPubMed Welle S, Brooks AI, Delehanty JM, Needler N, Bhatt K, Shah B, et al. Skeletal muscle gene expression profiles in 20–29 year old and 65–71 year old women. Exp Gerontol. 2004;39:369–77.CrossRefPubMed
86.
87.
Zurück zum Zitat Keller P, Vollaard NB, Gustafsson T, Gallagher IJ, Sundberg CJ, Rankinen T, et al. A transcriptional map of the impact of endurance exercise training on skeletal muscle phenotype. J Appl Physiol. 2011;110:46–59.CrossRefPubMed Keller P, Vollaard NB, Gustafsson T, Gallagher IJ, Sundberg CJ, Rankinen T, et al. A transcriptional map of the impact of endurance exercise training on skeletal muscle phenotype. J Appl Physiol. 2011;110:46–59.CrossRefPubMed
88.
Zurück zum Zitat Debigare R, Maltais F, Cote CH, Michaud A, Caron MA, Mofarrahi M, et al. Profiling of mRNA expression in quadriceps of patients with COPD and muscle wasting. Copd. 2008;5:75–84.CrossRefPubMed Debigare R, Maltais F, Cote CH, Michaud A, Caron MA, Mofarrahi M, et al. Profiling of mRNA expression in quadriceps of patients with COPD and muscle wasting. Copd. 2008;5:75–84.CrossRefPubMed
89.
Zurück zum Zitat Crul T, Testelmans D, Spruit MA, Troosters T, Gosselink R, Geeraerts I, et al. Gene expression profiling in vastus lateralis muscle during an acute exacerbation of COPD. Cell Physiol Biochem. 2010;25:491–500.CrossRefPubMed Crul T, Testelmans D, Spruit MA, Troosters T, Gosselink R, Geeraerts I, et al. Gene expression profiling in vastus lateralis muscle during an acute exacerbation of COPD. Cell Physiol Biochem. 2010;25:491–500.CrossRefPubMed
90.
Zurück zum Zitat Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, et al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature. 1997;390:45–51.CrossRefPubMed Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, et al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature. 1997;390:45–51.CrossRefPubMed
91.
Zurück zum Zitat Kim JS, Kim EJ, Kim HJ, Yang JY, Hwang GS, Kim CW. Proteomic and metabolomic analysis of H2O2-induced premature senescent human mesenchymal stem cells. Exp Gerontol. 2011;46:500–10.CrossRefPubMed Kim JS, Kim EJ, Kim HJ, Yang JY, Hwang GS, Kim CW. Proteomic and metabolomic analysis of H2O2-induced premature senescent human mesenchymal stem cells. Exp Gerontol. 2011;46:500–10.CrossRefPubMed
92.
Zurück zum Zitat Janssen I, Heymsfield SB, Ross R. Low relative skeletal muscle mass (sarcopenia) in older persons is associated with functional impairment and physical disability. J Am Geriatr Soc. 2002;50:889–96.CrossRefPubMed Janssen I, Heymsfield SB, Ross R. Low relative skeletal muscle mass (sarcopenia) in older persons is associated with functional impairment and physical disability. J Am Geriatr Soc. 2002;50:889–96.CrossRefPubMed
93.
Zurück zum Zitat Schols AM, Wouters EF, Soeters PB, Westerterp KR. Body composition by bioelectrical-impedance analysis compared with deuterium dilution and skinfold anthropometry in patients with chronic obstructive pulmonary disease. Am J Clin Nutr. 1991;53:421–4.PubMed Schols AM, Wouters EF, Soeters PB, Westerterp KR. Body composition by bioelectrical-impedance analysis compared with deuterium dilution and skinfold anthropometry in patients with chronic obstructive pulmonary disease. Am J Clin Nutr. 1991;53:421–4.PubMed
94.
Zurück zum Zitat Bosaeus I, Wilcox G, Rothenberg E, Strauss BJ. Skeletal muscle mass in hospitalized elderly patients: Comparison of measurements by single-frequency BIA and DXA. Clin Nutr. 2014;33:426–31.CrossRefPubMed Bosaeus I, Wilcox G, Rothenberg E, Strauss BJ. Skeletal muscle mass in hospitalized elderly patients: Comparison of measurements by single-frequency BIA and DXA. Clin Nutr. 2014;33:426–31.CrossRefPubMed
95.
Zurück zum Zitat Seymour JM, Ward K, Sidhu PS, Puthucheary Z, Steier J, Jolley CJ, et al. Ultrasound measurement of rectus femoris cross-sectional area and the relationship with quadriceps strength in COPD. Thorax. 2009;64:418–23.CrossRefPubMed Seymour JM, Ward K, Sidhu PS, Puthucheary Z, Steier J, Jolley CJ, et al. Ultrasound measurement of rectus femoris cross-sectional area and the relationship with quadriceps strength in COPD. Thorax. 2009;64:418–23.CrossRefPubMed
96.
Zurück zum Zitat Franssen FM, Broekhuizen R, Janssen PP, Wouters EF, Schols AM. Effects of whole-body exercise training on body composition and functional capacity in normal-weight patients with COPD. Chest. 2004;125:2021–8.CrossRefPubMed Franssen FM, Broekhuizen R, Janssen PP, Wouters EF, Schols AM. Effects of whole-body exercise training on body composition and functional capacity in normal-weight patients with COPD. Chest. 2004;125:2021–8.CrossRefPubMed
97.
Zurück zum Zitat Slinde F, Gronberg A, Engstrom CP, Rossander-Hulthen L, Larsson S. Body composition by bioelectrical impedance predicts mortality in chronic obstructive pulmonary disease patients. Respir Med. 2005;99:1004–9.CrossRefPubMed Slinde F, Gronberg A, Engstrom CP, Rossander-Hulthen L, Larsson S. Body composition by bioelectrical impedance predicts mortality in chronic obstructive pulmonary disease patients. Respir Med. 2005;99:1004–9.CrossRefPubMed
98.
Zurück zum Zitat Thibault R, Le Gallic E, Picard-Kossovsky M, Darmaun D, Chambellan A. [Assessment of nutritional status and body composition in patients with COPD: comparison of several methods]. Rev Mal Respir. 2010;27:693–702.CrossRefPubMed Thibault R, Le Gallic E, Picard-Kossovsky M, Darmaun D, Chambellan A. [Assessment of nutritional status and body composition in patients with COPD: comparison of several methods]. Rev Mal Respir. 2010;27:693–702.CrossRefPubMed
99.
Zurück zum Zitat Montes De Oca M, Loeb E, Torres SH, De Sanctis J, Hernandez N, Talamo C. Peripheral muscle alterations in non-COPD smokers. Chest. 2008;133:13–8.CrossRefPubMed Montes De Oca M, Loeb E, Torres SH, De Sanctis J, Hernandez N, Talamo C. Peripheral muscle alterations in non-COPD smokers. Chest. 2008;133:13–8.CrossRefPubMed
100.
Zurück zum Zitat Doll R, Peto R, Boreham J, Sutherland I. Mortality in relation to smoking: 50 years’ observations on male British doctors. BMJ. 2004;328:1519.CrossRefPubMedPubMedCentral Doll R, Peto R, Boreham J, Sutherland I. Mortality in relation to smoking: 50 years’ observations on male British doctors. BMJ. 2004;328:1519.CrossRefPubMedPubMedCentral
Metadaten
Titel
Genome-wide mRNA expression profiling in vastus lateralis of COPD patients with low and normal fat free mass index and healthy controls
verfasst von
Roberto A Rabinovich
Ellen Drost
Jonathan R Manning
Donald R Dunbar
MaCarmen Díaz-Ramos
Ramzi Lakhdar
Ricardo Bastos
William MacNee
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2015
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-014-0139-5

Weitere Artikel der Ausgabe 1/2015

Respiratory Research 1/2015 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Reizdarmsyndrom: Diäten wirksamer als Medikamente

29.04.2024 Reizdarmsyndrom Nachrichten

Bei Reizdarmsyndrom scheinen Diäten, wie etwa die FODMAP-arme oder die kohlenhydratreduzierte Ernährung, effektiver als eine medikamentöse Therapie zu sein. Das hat eine Studie aus Schweden ergeben, die die drei Therapieoptionen im direkten Vergleich analysierte.

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.

Update Innere Medizin

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