Skip to main content
Erschienen in: Arthritis Research & Therapy 1/2016

Open Access 01.12.2016 | Research article

Effects on muscle tissue remodeling and lipid metabolism in muscle tissue from adult patients with polymyositis or dermatomyositis treated with immunosuppressive agents

verfasst von: Ingela Loell, Joan Raouf, Yi-Wen Chen, Rongye Shi, Inger Nennesmo, Helene Alexanderson, Maryam Dastmalchi, Kanneboyina Nagaraju, Marina Korotkova, Ingrid E. Lundberg

Erschienen in: Arthritis Research & Therapy | Ausgabe 1/2016

Abstract

Background

Polymyositis (PM) and dermatomyositis (DM) are autoimmune muscle diseases, conventionally treated with high doses of glucocorticoids in combination with immunosuppressive drugs. Treatment is often dissatisfying, with persisting muscle impairment. We aimed to investigate molecular mechanisms that might contribute to the persisting muscle impairment despite immunosuppressive treatment in adult patients with PM or DM using gene expression profiling of repeated muscle biopsies.

Methods

Paired skeletal muscle biopsies from six newly diagnosed adult patients with DM or PM taken before and after conventional immunosuppressive treatment were examined by gene expression microarray analysis. Selected genes that displayed changes in expression were analyzed by Western blot. Muscle biopsy sections were evaluated for inflammation, T lymphocytes (CD3), macrophages (CD68), major histocompatibility complex (MHC) class I expression and fiber type composition.

Results

After treatment, genes related to immune response and inflammation, including inflammasome pathways and interferon, were downregulated. This was confirmed at the protein level for AIM-2 and caspase-1 in the inflammasome pathway. Changes in genes involved in muscle tissue remodeling suggested a negative effect on muscle regeneration and growth. Gene markers for fast type II fibers were upregulated and fiber composition was switched towards type II fibers in response to treatment. The expression of genes involved in lipid metabolism was altered, suggesting a potential lipotoxic effect on muscles of the immunosuppressive treatment.

Conclusion

The anti-inflammatory effect of immunosuppressive treatment was combined with negative effects on genes involved in muscle tissue remodeling and lipid metabolism, suggesting a negative effect on recovery of muscle performance which may contribute to persisting muscle impairment in adult patients with DM and PM.

Background

Polymyositis (PM) and dermatomyositis (DM) are chronic, idiopathic inflammatory myopathies (IIM) characterized by proximal muscle weakness. Muscle biopsies reveal signs of inflammation including infiltrating T cells, macrophages, cytokines (interleukin (IL)-1) interferons (IFNs)) and upregulated major histocompatibility complex (MHC) class I expression in the fibers as well as regenerating and degenerating fibers [1, 2]. Treatment is based on high doses of glucocorticoids (GC) often combined with additional immunosuppressive drugs. The effectiveness of GC in patients with PM or DM varies between individuals, but is often disappointing and few recover former muscle performance [35]. In addition, side effects such as osteoporosis, hypertension, insulin resistance and steroid myopathy are common [6].
GC interact with the glucocorticoid receptor (GR) and form a complex that is translocated into the cell nucleus where it regulates target gene actions through transrepression or transactivation mechanisms [79]. It is assumed that the immunosuppressive and anti-inflammatory effects of GC are mediated through transrepression, downregulating the expression of pro-inflammatory cytokines such as IL-1, tumor necrosis factor (TNF) and IFNγ [9]. On the other hand, transactivation through GC response elements (GREs) controls genes that mediate metabolic side effects of GC and enhances the expression of anti-inflammatory genes such as IL-10, IKB and annexin-1 [9]. The limited effects of conventional immunosuppressive treatment, including high doses of GC, on muscle performance in patients with PM and DM is well recognized, but the underlying molecular mechanisms of the limited effects have not been completely elucidated. Persisting upregulation of certain inflammatory pathways such as infiltrating T cells, MHC-I, several pro-inflammatory cytokines [1012], prostaglandin E2 (PGE2) [13] and leukotriene B4 (LTB4) pathways [14] in muscle tissue might partly explain the sustained weakness in patients despite treatment. Other molecular mechanisms affected by treatment may also influence muscle performance. This emphasizes the need for a better understanding of the molecular response in the target organ (muscle) in order to identify new therapeutic targets and abolish the persistent muscle weakness.
In this study, we aimed to investigate molecular events that might contribute to persisting compromised muscle function despite immunosuppressive treatment in adult patients with PM or DM. Thus, we investigated muscle biopsies taken before and after conventional immunosuppressive treatment using gene expression profiling combined with analysis of selected proteins at the protein level.

Methods

Patients and muscle biopsies

From an observational study, six untreated adult patients of Caucasian origin diagnosed with probable or definite DM or PM [15] were all subject to follow-up biopsies for the study. Disease duration was defined from the first reported symptom related to disease to time of the first muscle biopsy. Clinical data including support for diagnosis are presented in Table 1. All adult patients were initially treated with oral prednisolone (0.75 mg/kg/day) in combination with an additional immunosuppressive drug (methotrexate or azathioprine) as decided by the treating physician. Muscle tissue biopsies were taken from m. vastus lateralis; a repeated biopsy was taken after 9 months (range 8–15 months) of conventional immunosuppressive treatment [16]. None of the patients exercised at the time of the first biopsy, but all were instructed to a 5-days-a-week home exercise program after introduction of glucocorticoids. Patients one, two, four, and five exercised regularly with the home exercise program or more intensive gym training 1–2 times a week during the study period. The regional ethics committee in Stockholm granted approval (approval number: 2005/792-31/4) and all participants gave informed consent to participate in the study.
Table 1
Clinical data on the patients at the time of biopsies
Patient ID
Diagnosis
Age (years)
Gender
Disease duration (months)
Cumulative cortisone (mg)
Imuunosuppressive treatment at second biopsy
Autoantibodies
Support for diagnosis
MMT-8 (%)
s-CK (μcat/L)
HAQ (0.0–3.0)
FI-2 (%)
B
A
B
A
B
A
B
A
1
DM
40
M
3
14450.0
Pred, AZA
MDA5, SSA, Ro60
MW, S, LD
87.5
87.5
1.3
1.1
1.13
0.75
21.7
27.4
2
PM
73
F
12
6222.5
Pred, MTX
Neg
MW, CK, MB
85.0
85.0
10.2
1.5
0.5
0.88
18.3
19.1
3
DM
69
F
2
10017.5
Pred, MTX
ANA, TIF1γ
MW, CK, S, EMG
81.4
91.3
29.3
1.0
0.63
0
0
11.2
4
DM
63
M
0.5
7330.0
Pred, AZA
Neg
MW, CK, S, EMG
78.8
90.0
48.2
1.0
0,38
0
86.2
100.0
5
DM
45
F
16
6842.5
Pred, MTX
ANA, Mi-2
MW, CK, MB, S, EMG
98.8
87.5
43.0
2.1
1.5
0.88
17.3
25.3
6
PM
71
F
12
8415.0
Pred, AZA
Neg
MW, CK, MB
75.7
100
9.0
3.9
1.25
1.88
NA
17
A after treatment, ANA antinuclear antibodies, AZA azathioprine, B before treatment, CK creatine kinase (reference interval, male: 3.3 μkat/L, female: 2.5 μkat/L), DM dermatomyositis, EMG positive for electromyography, F female, FI-2 Functional Index-2 (0–100 %; impairment in performing repetitions, respective no impairment), HAQ Health Assessment Questionnaire (0.00–3.00; no impairment, respective impairment), LD lactate dehydrogenase (reference interval 105–333 IU/L), M male, MB positive muscle biopsy, MDA5 melanoma differentiation-associated protein 5, MMT-8 manual muscle testing in 8 muscle groups (0–100 %; muscle strength), MTX methotrexate, MW muscle weakness, NA not available, PM polymyositis, Pred prednisone, S skin rash, s-CK serum creatine kinase, SSA anti-Sjögren’s syndrome-related antigen A (also called anti-Ro), TIF1γ transcription intermediary factor 1-gamma

Clinical and laboratory assessment

Clinical and laboratory outcome measures were retrieved from the SweMyoNet quality of care register for myositis patients and from medical records. Muscle performance before and after treatment was assessed by the Manual Muscle Test (MMT-8) and the Functional Index-2 (FI-2); ≥15 % increase was defined as improved [17]. The MMT-8 measures isometric muscle strength in eight muscle groups [18] and the FI-2 measures dynamic repetitive muscle performance; it includes seven muscle groups with a maximum of 60 or 120 repetitions for each muscle [19]. Both the MMT-8 and the FI-2 are presented as % of maximal score (100 % = good muscle performance) in Table 1. Serum levels of creatine kinase (CK) and lactate dehydrogenase (LD) were analyzed as routine tests at the Department of Clinical Chemistry, Karolinska University Hospital. Myositis-associated and myositis-specific autoantibodies were tested by RNA immunoprecipitation (IP) and protein IP in Kyoto, Japan, and are presented in Table 1 [20, 21].

Histopathological and immunohistochemical analyses

Histopathological evaluation of muscle tissue sections was performed by an experienced muscle pathologist on coded sections stained with hematoxylin and eosin. Immunohistochemistry staining was used to identify the presence of inflammatory cells such as T lymphocytes (CD3), macrophages (CD68) and the expression of MHC class I according to a standard protocol [22] using mouse monoclonal anti-CD3 (BD Biosciences, CA, USA), anti-CD68 (Dako Cytomation, Denmark) and anti-MHC-I (My Bio Sourse, CA, USA) antibodies. Isotype-matched irrelevant antibodies were used as negative controls. Conventional microscopic evaluation of the staining was performed and the whole tissue sections were scored for CD3 and CD68 as follows: 0, no positive cells; 1, few positive scattered cells or one infiltrate of inflammatory cells; 2, clusters of positive cells or two infiltrates of inflammatory cells; and 3, several large cellular infiltrates. For MHC-I staining, the sections were scored as follows: 0, no positive fibers; 1, few positive scattered fibers; 2, clusters of positive fibers; and 3, several large areas with positive fibers.
Fiber-type composition was determined by mATPase staining to distinguish between slow-twitch type I and fast-twitch type II muscle fibers [23, 24]. In brief, muscle sections were pre-incubated at acidic or alkaline pH, respectively. Type I fibers emerge in a black color at pH 4.3 in contrast to type II fibers which appears in white; the opposite pattern is observed when pre-incubating at pH 10.3. Semi-quantitative analysis was applied on coded sections for analysis of fiber-type composition; the whole tissue section area was evaluated by counting fibers using a Leica microscope system (BX60; digital camera, Sony CDK-500, Tokyo, Japan). The results are presented as fiber type percentage of the total amount of fibers on the section.

RNA expression profiling

Expression profiling was performed using Affymetrix Human Genome U133 Plus 2.0 microarrays. Total RNA isolation, cDNA synthesis, cRNA labeling, microarray hybridization, and image acquisition were performed according to the manufacturer’s protocol [25]. The quality control criteria developed at the Children’s National Medical Center Microarray Center for each array were followed [25].
Hybridization signals of the microarrays were recorded using Microarray Suite 5.0 (MAS 5.0) (Affymetrix) and the data were analyzed using GeneSpring 7.0 (Agilent, CA, USA). Genes were filtered with the number of present calls across the 12 arrays analyzed. Genes with at least one present call were selected for statistical analysis using paired t test. All profiles have been made publicly accessible via NCBI GEO (http://​www.​ncbi.​nlm.​nih.​gov/​geo/​).
Genes with a fold change ≥2 were selected, and a functional analysis of the molecular networks and pathways was performed using the Ingenuity Pathway Analysis (IPA; Ingenuity Systems®, www.​ingenuity.​com). The significance of the association between the genes in the dataset, biological functions, and pathways was determined by the right-tailed Fischer’s exact test.

Western blot

Western blot was performed by using a tissue section protocol [26]. The 10-μm muscle sections were lysed in Tissue Protein Extraction Reagent (T-PER; Thermo Scientifics, USA) supplemented with 1× complete protease inhibitor cocktail (Roche Diagnostics GmbH, Mannheim, Germany) and incubated on ice for 30 min. The protein content was determined using a Bio-Rad protein assay (Bio-Rad Laboratories AB, Sweden). Gel electrophoresis was carried out on the NuPAGE® Novex® Bis-Tris gel system (Invitrogen AB, Sweden). Proteins were transferred on a polyvinylidene difluoride membrane using a Trans-Blot SD semi-dry transfer cell (Bio-Rad Laboratories). The membrane was blocked with 5 % milk in phosphate-buffered saline (PBS; 0.1 % Tween-20) and incubated with primary (rabbit polyclonal anti-caspase-1 (Millipore, MA, USA), rabbit polyclonal anti-FKBP5 (Millipore, MA, USA), mouse monoclonal anti-AIM-2 (LifeSpan Biosciences, WA, USA)) (overnight, 4 °C) and secondary (ECL anti-mouse IgG HRP linked (GE Healthcare, UK), ECL anti-rabbit IgG HRP linked (GE Healthcare, UK)) (1 h, room temperature) antibodies. The bands were detected by enhanced chemiluminescence (ECL) and the band intensities were measured using the Gel Doc XR system (Bio-Rad Laboratories). Quantification was performed with normalization against GAPDH as a housekeeping protein.

Statistical analyses

Clinical and experimental data were analyzed using Wilcoxon signed rank test. The level of significance was set at a p value ≤0.05.

Results

Effects of treatment on clinical parameters

Clinical data are summarized in Table 1. All untreated patients had a median of 7.5 months (range 0.5–16 months) duration of clinical symptoms to the first biopsy, which was taken as part of the diagnostic work-up. At the time of the second biopsy, after a median of 9 months (range 8–15 months) with immunosuppressive treatment, two out of six adult patients fulfilled the definition of improvement for MMT-8, and four patients improved for FI-2. One out of the six patients achieved the maximum score of 100 % but still had a low test on endurance FI-2, and only one reached the maximum test of FI-2 at the second biopsy, indicating persisting muscle impartment in almost all patients (Table 1). All patients had normal CK values at the second biopsy (Table 1).

Histopathological and immunohistochemical changes in pre- and post- treatment muscle biopsies

In the pre-treatment biopsy, four patients had detectable inflammatory cells: two had large inflammatory infiltrates, and two had scattered T lymphocytes or macrophages. Five out of the six patients had detectable positive staining for MHC-I expression in muscle fiber membranes, ranging from small areas with discrete staining to large areas with whole fibers expressing MHC-I. In the follow-up biopsy after immunosuppressive treatment, a few scattered T lymphocytes and macrophages were present in one patient, and scattered T lymphocytes were found in another patient. MHC class I expression was expressed in muscle fibers in one of five available follow-up biopsies. In addition, two pre-treatment biopsies showed signs of degenerating or regenerating fibers, but none of the follow-up biopsies showed this.

Effects of treatment on the overall gene expression

After treatment, the expression of 369 genes was significantly affected (>2.0 fold change) in the muscle tissue of patients, including 126 upregulated and 243 downregulated genes. Gene Ontology analysis demonstrated that the top Upstream Regulators statistically relevant for our gene dataset were Interferon Gamma (IFNG), interferon regulatory factor 7 (IRF7), Interferon type I (IFNα), signal transducer and activator of transcription 2 (STAT2) and Interferon Alfa 2 (IFNA2), which were predicted to be inhibited based on the gene expression changes in the dataset.

Effects on genes associated with immune response and inflammation

Gene Ontology analysis showed that the expression of 39 out of 43 genes associated with immune response and inflammation was downregulated by treatment (Table 2). Among the downregulated genes, a high representation of HLA-genes encoding MHC-I and MHC-II (which present antigens to CD8+ and CD4+ T cells, respectively) was seen. The expression of the co-stimulatory molecules CD80 and CD86 was also reduced. Moreover, a variety of chemokine receptors and ligands, both α- and β-chemokines, were downregulated (Table 2). Furthermore, the interferon signaling pathway was strongly downregulated in response to treatment. The expression of 13 genes, which are involved in type I as well as in type II IFN signaling, was reduced (Table 2, Fig. 1). Moreover, Absent in melanoma 2 (AIM2) and Caspase-1 (CASP1), components of an inflammasome complex promoting inflammation, were also downregulated. Additionally, specific receptors for pro-inflammatory lipid mediators such as Prostaglandin E Receptor 4 (PTGER4) and Cysteinyl Leukotriene Receptor 1 (CYSLTR1) were downregulated by treatment.
Table 2
Changes in expression (cutoff 2-fold) of the genes involved in immune responses and inflammation in patients with polymyositis or dermatomyositis after a median of 8.5 months of immunosuppressive treatment
Gene symbol
Gene
Affy #
Fold change
p
Immune response and antigen presentation
 
 CCL2
chemokine (C-C motif) ligand 2
216598_s_at
–5.9
0.004
 CCL5
chemokine (C-C motif) ligand 5
1405_i_at
–3.0
0.043
 CCR2
chemokine (C-C motif) receptor 2
206978_at
–2.3
0.004
 CCR5
chemokine (C-C motif) receptor 5
206991_s_at
–2.8
0.027
 CD52
CDW52 antigen (CAMPATH-1 antigen)
204661_at
–2.7
0.037
 CD80
CD80 antigen (CD28 ag ligand 1, B7-1 ag)
1554519_at
–2.2
0.034
 CD86
CD86 antigen (CD28 ag ligand 2, B7-2 ag)
210895_s_at
–2.6
0.013
 CHRNA1
cholinergic receptor, nicotinic, αpolypeptide 1
206633_at
–2.8
0.028
 CNPY3
trinucleotide repeat containing 5
1556389_at
–2.1
0.022
 CPM
carboxypeptidase M
206100_at
2.2
0.028
 HLA-DQB1
MHC class II, DQβ2
212998_x_at
–2.0
0.033
 HLA-A
major histocompatibility complex, class I, A
215313_x_at
–2.2
0.012
 HLA-G
HLA-G histocompatibility antigen, class I, G
211530_x_at
–2.3
0.010
 HLA-C
MHC class I, C
208812_x_at
–2.2
0.013
 HLA-B
MHC class I, B
209140_x_at
–2.2
0.017
 HLA-F
MHC class I, F
204806_x_at
–2.6
0.018
 HLA-DQA1
MHC class II, DQα1
203290_at
–2.6
0.036
 HLA-DQB1
MHC class II, DQβ1
209823_x_at
–2.8
0.010
 HLA-DPA1
MHC class II, DPα1
213537_at
–2.9
0.009
 IL-23A
Interleukin 23, subunit alpha
217328_at
–5.2
0.005
 IL-12RB1
Interleukin 12 receptor, beta 1
1552584_at
–2.1
0.020
 NMU
Neuromedin U
206023_at
2.8
0.028
 MMP3
Matrix metalloproteinase 3
205828_at
10.7
0.023
IFN pathway
 STAT1
signal transducer & activator of transcription 1, 91 kDa
209969_s_at
–3.3
0.008
 CXCL10
chemokine (C-X-C motif) ligand 10
204533_at
–5.6
0.020
 CXCL11
chemokine (C-X-C motif) ligand 11
211122_s_at
–5.6
0.015
 RTP4
28kD interferon responsive protein
219684_at
–5.7
0.028
 IRF8
IFN consensus sequence binding protein 1
204057_at
–2.4
0.033
 ISG20
IFN stimulated gene 20 kDa
204698_at
–5.8
0.029
 IFI6
IFNα-inducible protein
204415_at
–4.7
0.045
 IFI30
IFNγ-inducible protein 30
201422_at
–2.2
0.036
 IFI35
IFN -induced protein 35
209417_s_at
–2.6
0.036
 IFIT3
IFN -induced protein w tetratricopeptide repeats 4
229450_at
–5.1
0.032
 IRF9
IFN -stimulated transcription factor 3, γ
203882_at
–3.8
0.005
 GBP1
guanylate binding protein 1, IFN-inducible
202269_x_at
–2.9
0.009
 GBP2
guanylate binding protein 1, IFN-inducible
242907_at
–2.7
0.005
 GBP5
guanylate binding protein 5
238581_at
–2.1
0.017
Inflammasome
 AIM2
absent in melanoma 2
206513_at
–2.5
0.008
 CASP1
caspase 1, (interleukin 1β convertase)
211367_s_at
–2.3
0.009
 IL18
interleukin 18 (IFNg-inducing factor)
206295_at
–2.2
0.042
Eicosanoids
 PTGER3
prostaglandin E receptor 3 (subtype EP3)
210832_x_at
3.0
0.013
 PTGER4
prostaglandin E receptor 4 (subtype EP4)
204897_at
–2.0
0.027
 CYSLTR1
cysteinyl leukotriene receptor 1
230866_at
–2.8
0.037

Effects on genes involved in muscle tissue remodeling

A number of genes associated with muscle tissue remodeling were affected by treatment (Table 3). Five genes associated with the ubiquitin-proteasome pathway were downregulated. The GR co-chaperone protein FK506 binding protein 5 (FKBP5) was upregulated while Nuclear receptor co-activator 6 (NCOA6) was decreased after treatment. The expression of the genes for sarcomeric muscle protein α-actinin 3 (ACTN3) and vinculin (VCL) was enhanced, suggesting a compensatory increase to cope with the muscle loss due to degeneration. However, the negative regulator of muscle growth Myostatin (MSTN) was also upregulated suggesting active inhibition of muscle growth, while Bone morphologic protein 1 (BMP1) protease that can regulate MSTN by cleaving was downregulated after treatment. Also, Ras associated with diabetes (RRAD) and Myosin binding protein H (MYBPH) that is involved early in skeletal muscle development was suppressed upon treatment suggesting reduced fiber regeneration. IPA functional analysis based on over-representation and expression direction of genes in our data set predicted the size of muscle cells and development of blood vessels to be reduced after treatment (Z-score –2.108 and –2.509, respectively). These data indicate negative effects of treatment on muscle fiber differentiation and growth. In addition, the expression of the myosin heavy chain 4 (MYH4) and ACTN3, specific markers for fast type II fibers, was upregulated suggesting a fiber type switch towards fast type II fibers in response to treatment.
Table 3
Changes in expression (cutoff 2-fold) of genes involved in ubiquitin proteasome pathway, skeletal muscle structure, and remodeling in patients with polymyositis or dermatomyositis after immunosuppressive treatment
Gene symbol
Gene
Affy #
Fold change
p
Ubiquitin proteasome pathway
 PSMB8
proteasome subunit,β type, 8 (large multifunctional protease 7)
209040_s_at
–6.6
0.003
 UBE2L6
ubiquitin-conjugating enzyme E2L 6
201649_at
–2.8
0.032
 PSMB9
proteasome (prosome, macropain) subunit, beta type, 9
204279_at
–2.4
0.005
 PSME1
proteasome) activator subunit 1 (PA28 α)
200814_at
–2.3
0.007
 PSME2
proteasome activator subunit 2 (PA28β)
201762_s_at
–2.0
0.012
 CNTN3
ubiquitin-activating enzyme E1C (UBA3 homolog, yeast)
229831_at
2.4
0.029
Structure proteins and tissue remodeling
 MYBPH
myosin binding protein H
206304_at
–6.9
0.036
 RRAD
Ras-related associated with diabetes
204803_s_at
–3.2
0.013
 BMP1
bone morphogenetic protein 1
207595_s_at
–2.7
0.001
 NCOA6
Nuclear receptor co-activator
1568874_at
–3.0
0.041
 CACNA1D
calcium channel, voltage-dependent, L type, alpha 1D subunit
1555993_at
–2.9
0.035
 CHST11
carbohydrate (chondroitin 4) sulfotransferase 11
226368_at
–2.1
0.009
 MYH4
myosin, heavy polypeptide 4, skeletal muscle
208148_at
2.2
0.020
 FOXO1
forkhead box O1A
202723_s_at
2.3
0.026
 MSTN
growth differentiation factor 8
207145_at
2.3
0.041
 VCL
vinculin
200930_s_at
2.4
0.022
 TIMP4
tissue inhibitor of metalloproteinase 4
206243_at
2.6
0.024
 FKBP5
FK506 binding protein 5
204560_at
3.4
0.015
 ACTN3
actinin, alpha 3
206891_at
3.4
0.037

Effects on genes involved in lipid metabolism

Treatment resulted in significant changes in the expression of genes involved in lipid metabolism (Table 4). Genes responsible for fatty acid (FA) uptake and transport such as fatty acid binding protein 7 (FABP7) and ATP-binding cassette, sub-family D member 2 (ABCD2) were upregulated. Moreover, genes that promote lipolysis such as Lipoprotein Lipase (LPL), Hormone-sensitive lipase (LIPE), and Carboxylesterase 1 (CES1) were also upregulated, while the genes that protect from lipolysis, for instance Lipid Storage Droplet Protein (LSDP5), were suppressed suggesting enhanced generation of free FA. Genes associated with FA oxidation and oxidative phosphorylation was not affected (data not shown), suggesting partition of FA into intramuscular lipids. Moreover, genes that favor lipogenesis and lipid storage, e.g. stearoyl-CoA desaturase (delta-9) (SCD), cell death-inducing DFFA-like effector c (CIDEC), and ceramide synthase 3 (CERS3), were enhanced (Table 4). In line with these results, based on expression results of genes in the data set, storage of lipids was predicted to be increased (Z-score +2.066). Notably, the expression of sphingosine kinase 1 (SPHK1) was decreased, suggesting enhanced accumulation of ceramide, an important lipid mediator previously implicated in lipotoxicity [27].
Table 4
Changes in expression of the genes involved in lipid metabolism in patients with polymyositis or dermatomyositis after immunosuppressive treatment
Gene symbol
Gene
Affy #
Fold change
p
Lipid transport and uptake
 FABP7
fatty acid binding protein 7, brain
205029_s_at
10.0
0.002
 ABCD2
ATP-binding cassette, sub-family D member 2
207583_at
4.55
0.043
 APOL6
apolipoprotein L, 6
241869_at
–3.12
3.14E-05
Lipid accumulation and lipolysis
 SCD
stearoyl-CoA desaturase (delta-9)
223839_s_at
3.86
0.042
 CIDEC
cell death-inducing DFFA-like effector c
219398_at
3.53
0.049
 CERS3
ceramide synthase 3
1554252_a_at
3.2
0.021
 CES1
carboxylesterase 1
209616_s_at
2.98
0.028
 MSTN
myostatin
207145_at
2.28
0.041
 CNR1
Human CB1 cannabinoid receptor
213436_at
2.14
0.009
 LPL
lipoprotein lipase
203549_s_at
2.05
0.022
 LIPE
lipase, hormone-sensitive
213855_s_at
2.00
0.034
 ACSL3
fatty-acid-Coenzyme A ligase, long-chain 3
236168_at
–3.87
0.048
 LSDP5
Lipid Storage Droplet Protein 5
1560457_x_at
–2.61
0.030
 SPHK1
sphingosine kinase 1
219257_s_at
–2.20
0.037

Confirmation of changes at the protein level

To confirm changes in gene expression at the protein level by Western blot we selected eight genes that were significantly changed. Two of the chosen genes are involved in the inflammatory pathway (AIM-2 and Caspase-1), which were both downregulated after treatment. The FKBP5 gene is implicated in muscle tissue remodeling and was upregulated after treatment. Using Western blot, we confirmed significantly reduced protein expression of AIM-2 and Caspase-1 (p < 0.05), suggesting a reduction in inflammatory signaling (Fig. 2). We also observed an increased protein expression of FKBP5 (p < 0.05), supporting a negative effect on muscle tissue remodeling by the immunosuppressive treatment. No significant changes for EP3, EP4, CystLTR1, FOXO1A, and FABP7 were detected at the protein level (data not shown).

Effects on fiber type composition

A switch in fiber types was seen in the post-treatment biopsy as compared to that before treatment. The percentage of type I fibers had decreased significantly after treatment, from a median of 52 % (range 31–57 %) to 43 % (range 14–46 %) (p < 0.05). In contrast, the proportion of type II fibers was significantly higher after treatment (before treatment, median 48 % (range 43–69 %); after treatment, 57 % (54–86 %); p < 0.05), thus confirming the gene expression data (Fig. 3).

Discussion

In the present study, in which adult patients improved but none had recovered muscle strength at the follow-up biopsy, we found that immunosuppressive treatment of newly diagnosed PM and DM patients had suppressive effects on gene expression of immune and inflammatory pathways, including type 1 IFN and inflammasome pathways, in skeletal muscle. However, we also observed changed expression of genes involved in skeletal muscle tissue remodeling indicating protein breakdown and reduced muscle regeneration, which may negatively affect muscle regeneration and growth. Furthermore, we found altered expression of genes associated with lipid uptake, lipolysis, and lipid accumulation in response to treatment, indicating complex effects on intramuscular lipid metabolism that may also have a negative effect on muscle performance. Among the immune and inflammatory pathways suppressed by treatment, the downregulation of type I IFN pathways in muscle tissue was most striking. It is well recognized that the type I IFN pathway is activated in patients with autoimmune diseases including IIM [28, 29]. A significant upregulation of IFN-inducible genes in muscle biopsies from PM and DM patients was detected compared to age-/sex-matched controls [30, 31]. The high overexpression of interferon-inducible genes was also demonstrated in whole blood from both PM and DM patients [32]. Moreover, a recent study of peripheral blood gene expression has revealed that IIM patients displayed a predominant IFNα-mediated response program [29]. The expression of type I IFN-inducible genes in whole blood correlated with disease activity in PM and DM patients and was reduced after immunomodulatory therapies [32, 33]. Our novel finding that immunosuppressive treatment suppressed the IFN pathway in muscle tissue from PM and DM patients is in agreement with these previous reports. Our results provide additional evidence supporting the beneficial effects of conventional immunosuppressive treatment in myositis, through inhibition of the IFN pathway and reduced formation of pro-inflammatory mediators in muscle tissue.
Another finding was downregulation of genes involved in inflammasome activity in response to treatment, which was confirmed at the protein level for AIM-2 and Caspase-1. Our findings have added insights into the favorable effects of conventional immunosuppressive treatment, which includes inhibition of the inflammasome pathway in muscle tissue in patients with PM or DM, as well as several other pathways associated with immune response and inflammation, which was validated by immunohistochemistry confirming a low degree of inflammation in the post-treatment biopsies as assessed by CD3, CD68, and MHC-I expression.
However, our group has previously demonstrated an insufficient effect of immunosuppressive treatment on PGE2 and LTB4 pathways associated with the persistent expression of mPGES-1, COX-1, and 5-LO proteins in myositis muscle despite treatment [13, 14]. In line with these observations, we did not detect any alterations in the gene expression of these enzymes or changes at the protein level for the eicosanoid receptors EP3, EP4, and CysLTR1. The receptors were expressed at the protein level in muscle from patients with myositis before and after treatment, suggesting that PGE2 and LT might contribute to chronic inflammation and muscle wasting and these pathways could be potential targets for new therapies.
Importantly we found signs in the gene expression profiles after treatment indicating an effect on muscle remodeling. We observed downregulation of several genes in the ubiquitin-proteasome pathway and also increased expression of structural proteins such as α-actinin and vinculin, indicating an increase in muscle mass. Reversely, we detected increased expression of myostatin, suggesting inhibition of myogenesis and a negative effect on muscle growth. Furthermore, downregulation of RRAD and MYBPH could also be a sign of reduced muscle regeneration. RRAD expression was elevated during skeletal muscle development as well as in adult muscle post-injury [34]. FKBP5 is an essential functional regulator of the GR complex and is associated with muscle tissue alteration; it plays an important role in basic cellular processes and in immunoregulation involving protein folding and trafficking [35]. We observed an increased protein expression of FKBP5, implicating a negative effect on muscle tissue remodeling. Overall, these data point to negative effects of conventional immunosuppressive treatment on muscle regeneration and growth. Furthermore, the enhanced gene expression of specific markers for fast type II fibers, MYH4 and ACTN3, suggest a fiber-type switching towards the type II fibers in response to treatment, which was confirmed by analysis of fiber-type composition. This observation is in agreement with the clinical problem of low muscle endurance as measured by FI-2 and with previous data reporting a shift towards fast twitch type II fibers in patients with chronic PM or DM which interestingly could be reversed by exercise [36, 37].
A third pathway that we found to be altered in muscle tissue after immunosuppressive treatment relates to lipid metabolism. The balance between lipid production and oxidation is essential for normal cell functions; thus, an excess of FFA is converted to triacylglycerol for intracellular lipid storage. The dysregulation of this process leads to the production of lipotoxic lipid intermediates (ceramides, diacylglycerol, fatty acyl CoA) that might cause cell dysfunction or death [38]. A novel observation from our study is that immunosuppressive treatment including GC might affect lipid storage in skeletal muscle. In addition, upregulated CERS3 suggests an enhanced accumulation of ceramide which has previously been linked to insulin resistance [39]. Moreover, ceramide has been implicated in skeletal muscle dysfunction and fatigue in chronic diseases and in mouse muscle fibers in vitro [40, 41]. Additional detailed studies are needed to define lipid profiles in muscle tissue from myositis patients in comparison with healthy individuals and in relation to immunosuppressive treatment. Notably, patients with juvenile DM are at risk of developing lipodystrophy, associated with loss or redistribution of subcutaneous fat [42]. The lipodystrophy is accompanied by metabolic abnormalities such as insulin resistance, diabetes and dyslipidemia, and may occur as a result of inflammation. Our study included adult patients, although there is very little known about lipodystrophy in adult patients with PM or DM. There is a case study from 2007 describing a woman suffering from a typical DM which developed lipodystrophy and insulin resistance [43]. Although worth mentioning, there is no evidence that standard therapies for DM causes lipodystrophy.
A strength of our study is the paired muscle biopsy samples, with two biopsies taken from the same individuals and the repeated biopsy that was taken regardless of clinical signs of a flare. A paired sample study design reduces the problem of inter-individual variations. Nevertheless, our current study has several limitations: one of them is the low number of patients included and the heterogeneity in diagnoses of PM and DM and in the degree of histopathological changes before treatment. Also, no magnetic resonance imaging (MRI) was performed before the biopsies were taken which could have enhanced the detection of inflammation in the muscle. Differences in typical histopathological features in muscle biopsies seen in PM and DM suggest that different mechanisms may contribute to the muscle inflammation. However, several studies on cytokine and chemokine expression have not revealed significant differences between PM and DM, suggesting that inflammatory molecular pathways may be shared. One patient with typical DM features and muscle weakness had no signs of MHC class expression on muscle fibers, which could be explained by the sometimes patchy distribution of MHC class I expression. Another limitation is the inconsistency in the immunosuppressive treatment used in combination with GC, as it was given based on the decision of the treating physician, although all patients were treated with high doses of GC. Furthermore, the total expected duration of immunosuppressive treatment in patients with PM or DM is often 2–3 years. Here, we chose to take a repeated biopsy after approximately 9 months, which is not likely to show the final repaired muscle but rather an effect of the immunosuppressive treatment on molecular pathways (which was the aim of our study). Despite the heterogeneity in diagnosis and treatment and the low degree of inflammatory cell infiltrates in two patients before treatment, we could still see significant downregulation of genes involved in inflammation, supporting the beneficial effect of the immunosuppressive treatment on the inflammatory pathway. One patient developed type 2 diabetes after the start of immunosuppressive treatment. None of the other patients had medications or conditions that could impact muscle metabolism. Furthermore, details on diet were not included. In recent years, our group has shown that intensive exercise can have a positive influence on muscle health [44]. Four out of the six patients in the present study did exercise regularly, which might have counteracted some of the damage induced by the oral corticosteroid treatment. Moreover, it is not possible to distinguish between the relative contribution of the disease progress and the immunosuppressive treatment on the outcome in this study. To address this question an experimental model should be considered. Due to the limited number of patients the results need to be interpreted with some caution and need to be replicated in a larger cohort of patients.

Conclusions

In conclusion, a majority of genes involved in immune response were downregulated in muscle tissue from patients with PM or DM after conventional immunosuppressive treatment. In addition, genes involved in protein degradation and muscle regeneration were altered, indicating insufficient muscle tissue remodeling, and, finally, the expression of genes related to lipid metabolism was affected by treatment, suggesting intramuscular lipid accumulation leading to skeletal muscle dysfunction. These findings provide new plausible explanations for the persistent muscle weakness and fatigue observed in patients despite treatment, and diminished tissue inflammation, and at least some of these may be affected in a beneficial way by combining immunosuppressive treatment with physical exercise.

Abbreviations

ABCD2, ATP-binding cassette, sub-family D member 2; ACTN3, sarcomeric muscle protein α-actinin 3; AIM2, Absent In melanoma 2; BMP1, Bone morphologic protein 1; CASP1, Caspase-1; CD3, T lymphocytes; CD68, macrophages; CERS3, ceramide synthase 3; CES1, Carboxylesterase 1; CIDEC, cell death-inducing DFFA-like effector c; CK, creatine kinase; CYSLTR1, Cysteinyl Leukotriene Receptor 1; DM, dermatomyositis; ECL, enhanced chemiluminescence; FA, fatty acid; FABP7, fatty acid binding protein 7; FI-2, Functional Index-2; FKBP5, FK506 binding protein 5; GC, glucocorticoids; GR, glucocorticoid receptor; GRE, glucocorticoid response element; HAQ, Health Assessment Questionnaire; IFNA2, interferon alfa 2; IFNG, interferon gamma; IFN, interferon; IFNα, interferon type I; IIM, idiopathic inflammatory myopathies; IL, interleukin; IPA, Ingenuity Pathway Analysis; IRF7, interferon regulatory factor 7; LIPE, Hormone-sensitive lipase; LPL, Lipoprotein Lipase; LSDP5, Lipid Storage Droplet Protein; LTB4, leukotriene B4; MHC, major histocompatibility complex; MMT-8, Manual Muscle Test; MRI, magnetic resonance imaging; MSTN, Myostatin; MYBPH, Myosin binding protein H; MYH4, myosin heavy chain 4; NCOA6, nuclear receptor co-activator 6; PGE2, prostaglandin E2; PM, polymyositis; PTGER4, prostaglandin E Receptor 4; RRAD, Ras associated with diabetes; SCD, stearoyl-CoA desaturase; SPHK1, sphingosine kinase 1; STAT2, signal transducer and activator of transcription 2; TNF, tumor necrosis factor; VCL, vinculin

Acknowledgements

The authors would like to thank Eva Lindroos for exceptional handling and provision of muscle biopsy samples. Many thanks are also given to Nurse Christina Ottosson for excellent patient care and for providing clinical data from patients. We want to thank Professor Tsuneyo Mimori, Kyoto University Graduate School of Medicine, Japan, for providing us with results for myositis-associated and -specific autoantibodies.

Funding

This study was supported by grants from the Swedish Research Council, the Swedish Rheumatism Association, King Gustaf V 80 Year Foundation, Funds at the Karolinska Institutet and through the regional agreement on medical training and clinical research (ALF) between Stockholm County Council and Karolinska Institutet. KN was supported by National Institutes of Health grantss K26OD011171, R24HD050846-02, and P50AR060836, and United States Department of Defense grants W81XWH-05-1-0659, W81XWH-11-1-0782, W81XWH-11-1-0330, and W81XWH 10-1-0767.

Authors’ contributions

IL carried out the immunohistochemistry staining, and participated in drafting and revising the manuscript. JR carried out the immunoblots, contributed to analysis and interpretation of data, and participated in drafting, revising, and finalized the manuscript. YWC carried out the gene expression profiling and participated in revising the manuscript. RS carried out the gene expression profiling and helped to revise the manuscript. IN contributed to analysis and interpretations of histopathology and immunohistochemistry staining, and participated in revising the manuscript. HA participated in the design of the study and participated in revising the manuscript. MD participated in the design of the study and helped to revise the manuscript. KN participated in the design of the study and revising the manuscript. MK contributed to the interpretation of data, and participated in the design of the study and revising the manuscript. IEL participated in the design of the study and revising the manuscript. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Nagaraju K, Casciola-Rosen L, Lundberg I, Rawat R, Cutting S, Thapliyal R, et al. Activation of the endoplasmic reticulum stress response in autoimmune myositis: potential role in muscle fiber damage and dysfunction. Arthritis Rheum. 2005;52(6):1824–35.CrossRefPubMed Nagaraju K, Casciola-Rosen L, Lundberg I, Rawat R, Cutting S, Thapliyal R, et al. Activation of the endoplasmic reticulum stress response in autoimmune myositis: potential role in muscle fiber damage and dysfunction. Arthritis Rheum. 2005;52(6):1824–35.CrossRefPubMed
2.
Zurück zum Zitat Lundberg I, Ulfgren AK, Nyberg P, Andersson U, Klareskog L. Cytokine production in muscle tissue of patients with idiopathic inflammatory myopathies. Arthritis Rheum. 1997;40(5):865–874. Lundberg I, Ulfgren AK, Nyberg P, Andersson U, Klareskog L. Cytokine production in muscle tissue of patients with idiopathic inflammatory myopathies. Arthritis Rheum. 1997;40(5):865–874.
3.
Zurück zum Zitat Zong M, Lundberg IE. Pathogenesis, classification and treatment of inflammatory myopathies. Nat Rev Rheumatol. 2011;7(5):297–306. Zong M, Lundberg IE. Pathogenesis, classification and treatment of inflammatory myopathies. Nat Rev Rheumatol. 2011;7(5):297–306.
4.
Zurück zum Zitat Hengstman G, Van Den Hoogen F, Van Engelen B. Treatment of dermatomyositis and polymyositis with anti-tumor necrosis factor-α: long-term follow-up. Eur Neurol. 2004;52(1):61–63. Hengstman G, Van Den Hoogen F, Van Engelen B. Treatment of dermatomyositis and polymyositis with anti-tumor necrosis factor-α: long-term follow-up. Eur Neurol. 2004;52(1):61–63.
5.
Zurück zum Zitat Pandya JM, Loell I, Hossain MS, Zong M, Alexanderson H, Raghavan S, et al. Effects of conventional immunosuppressive treatment on CD244+ (CD28null) and FOXP3+ T cells in the inflamed muscle of patients with polymyositis and dermatomyositis. Arthritis Res Ther. 2016;18(1):1.CrossRef Pandya JM, Loell I, Hossain MS, Zong M, Alexanderson H, Raghavan S, et al. Effects of conventional immunosuppressive treatment on CD244+ (CD28null) and FOXP3+ T cells in the inflamed muscle of patients with polymyositis and dermatomyositis. Arthritis Res Ther. 2016;18(1):1.CrossRef
6.
Zurück zum Zitat Schäcke H, Döcke W-D, Asadullah K. Mechanisms involved in the side effects of glucocorticoids. Pharmacol Ther. 2002;96(1):23–43.CrossRefPubMed Schäcke H, Döcke W-D, Asadullah K. Mechanisms involved in the side effects of glucocorticoids. Pharmacol Ther. 2002;96(1):23–43.CrossRefPubMed
7.
Zurück zum Zitat Necela BM, Cidlowski JA. Mechanisms of glucocorticoid receptor action in noninflammatory and inflammatory cells. Proc Am Thorac Soc. 2004;1(3):239–46.CrossRefPubMed Necela BM, Cidlowski JA. Mechanisms of glucocorticoid receptor action in noninflammatory and inflammatory cells. Proc Am Thorac Soc. 2004;1(3):239–46.CrossRefPubMed
8.
Zurück zum Zitat Hayashi R, Wada H, Ito K, Adcock IM. Effects of glucocorticoids on gene transcription. Eur J Pharmacol. 2004;500(1):51–62.CrossRefPubMed Hayashi R, Wada H, Ito K, Adcock IM. Effects of glucocorticoids on gene transcription. Eur J Pharmacol. 2004;500(1):51–62.CrossRefPubMed
9.
Zurück zum Zitat Newton R, Holden NS. Separating transrepression and transactivation: a distressing divorce for the glucocorticoid receptor? Mol Pharmacol. 2007;72(4):799–809.CrossRefPubMed Newton R, Holden NS. Separating transrepression and transactivation: a distressing divorce for the glucocorticoid receptor? Mol Pharmacol. 2007;72(4):799–809.CrossRefPubMed
10.
Zurück zum Zitat Nyberg P, Wikman AL, Nennesmo I, Lundberg I. Increased expression of interleukin 1alpha and MHC class I in muscle tissue of patients with chronic, inactive polymyositis and dermatomyositis. J Rheumatol. 2000;27(4):940–8.PubMed Nyberg P, Wikman AL, Nennesmo I, Lundberg I. Increased expression of interleukin 1alpha and MHC class I in muscle tissue of patients with chronic, inactive polymyositis and dermatomyositis. J Rheumatol. 2000;27(4):940–8.PubMed
11.
Zurück zum Zitat Zong M, Loell I, Lindroos E, Nader GA, Alexanderson H, Hallengren CS, Borg K, Arnardottir S, McInnes IB, Lundberg IE. Effects of immunosuppressive treatment on interleukin-15 and interleukin-15 receptor α expression in muscle tissue of patients with polymyositis or dermatomyositis. Ann Rheum Dis. 2012. annrheumdis-2011–200495. Zong M, Loell I, Lindroos E, Nader GA, Alexanderson H, Hallengren CS, Borg K, Arnardottir S, McInnes IB, Lundberg IE. Effects of immunosuppressive treatment on interleukin-15 and interleukin-15 receptor α expression in muscle tissue of patients with polymyositis or dermatomyositis. Ann Rheum Dis. 2012. annrheumdis-2011–200495.
12.
Zurück zum Zitat Ulfgren AK, Grundtman C, Borg K, Alexanderson H, Andersson U, Harris HE, et al. Down-regulation of the aberrant expression of the inflammation mediator high mobility group box chromosomal protein 1 in muscle tissue of patients with polymyositis and dermatomyositis treated with corticosteroids. Arthritis Rheum. 2004;50(5):1586–94.CrossRefPubMed Ulfgren AK, Grundtman C, Borg K, Alexanderson H, Andersson U, Harris HE, et al. Down-regulation of the aberrant expression of the inflammation mediator high mobility group box chromosomal protein 1 in muscle tissue of patients with polymyositis and dermatomyositis treated with corticosteroids. Arthritis Rheum. 2004;50(5):1586–94.CrossRefPubMed
13.
Zurück zum Zitat Korotkova M, Helmers SB, Loell I, Alexanderson H, Grundtman C, Dorph C, et al. Effects of immunosuppressive treatment on microsomal prostaglandin E synthase 1 and cyclooxygenases expression in muscle tissue of patients with polymyositis or dermatomyositis. Ann Rheum Dis. 2008;67(11):1596–602.CrossRefPubMed Korotkova M, Helmers SB, Loell I, Alexanderson H, Grundtman C, Dorph C, et al. Effects of immunosuppressive treatment on microsomal prostaglandin E synthase 1 and cyclooxygenases expression in muscle tissue of patients with polymyositis or dermatomyositis. Ann Rheum Dis. 2008;67(11):1596–602.CrossRefPubMed
14.
Zurück zum Zitat Loell I, Munters LA, Pandya J, Zong M, Alexanderson H, Fasth AE, Hallengren CS, Rådmark O, Lundberg IE, Jakobsson P-J. Activated LTB4 pathway in muscle tissue of patients with polymyositis or dermatomyositis. Ann Rheum Dis. 2013;72(2):293–299. Loell I, Munters LA, Pandya J, Zong M, Alexanderson H, Fasth AE, Hallengren CS, Rådmark O, Lundberg IE, Jakobsson P-J. Activated LTB4 pathway in muscle tissue of patients with polymyositis or dermatomyositis. Ann Rheum Dis. 2013;72(2):293–299.
15.
Zurück zum Zitat Bohan A, Peter JB, Bowman RL, Pearson CM. A computer-assisted analysis of 153 patients with polymyositis and dermatomyositis. Medicine. 1977;56(4):255–86.CrossRefPubMed Bohan A, Peter JB, Bowman RL, Pearson CM. A computer-assisted analysis of 153 patients with polymyositis and dermatomyositis. Medicine. 1977;56(4):255–86.CrossRefPubMed
16.
Zurück zum Zitat Dorph C, Nennesmo I, Lundberg IE. Percutaneous conchotome muscle biopsy. A useful diagnostic and assessment tool. J Rheumatol. 2001;28(7):1591–9.PubMed Dorph C, Nennesmo I, Lundberg IE. Percutaneous conchotome muscle biopsy. A useful diagnostic and assessment tool. J Rheumatol. 2001;28(7):1591–9.PubMed
17.
18.
Zurück zum Zitat Rider LG, Koziol D, Giannini EH, Jain MS, Smith MR, Whitney‐Mahoney K, et al. Validation of manual muscle testing and a subset of eight muscles for adult and juvenile idiopathic inflammatory myopathies. Arthritis Care Res. 2010;62(4):465–72.CrossRef Rider LG, Koziol D, Giannini EH, Jain MS, Smith MR, Whitney‐Mahoney K, et al. Validation of manual muscle testing and a subset of eight muscles for adult and juvenile idiopathic inflammatory myopathies. Arthritis Care Res. 2010;62(4):465–72.CrossRef
19.
Zurück zum Zitat Alexanderson H, Broman L, Tollback A, Josefson A, Lundberg IE, Stenstrom CH. Functional index-2: validity and reliability of a disease-specific measure of impairment in patients with polymyositis and dermatomyositis. Arthritis Rheum. 2006;55(1):114–22.CrossRefPubMed Alexanderson H, Broman L, Tollback A, Josefson A, Lundberg IE, Stenstrom CH. Functional index-2: validity and reliability of a disease-specific measure of impairment in patients with polymyositis and dermatomyositis. Arthritis Rheum. 2006;55(1):114–22.CrossRefPubMed
20.
Zurück zum Zitat Forman MS, Nakamura M, Mimori T, Gelpi C, Hardin JA. Detection of antibodies to small nuclear ribonucleoproteins and small cytoplasmic ribonucleoproteins using unlabeled cell extracts. Arthritis Rheum. 1985;28(12):1356–61.CrossRefPubMed Forman MS, Nakamura M, Mimori T, Gelpi C, Hardin JA. Detection of antibodies to small nuclear ribonucleoproteins and small cytoplasmic ribonucleoproteins using unlabeled cell extracts. Arthritis Rheum. 1985;28(12):1356–61.CrossRefPubMed
21.
Zurück zum Zitat Nakashima R, Imura Y, Kobayashi S, Yukawa N, Yoshifuji H, Nojima T, et al. The RIG-I-like receptor IFIH1/MDA5 is a dermatomyositis-specific autoantigen identified by the anti-CADM-140 antibody. Rheumatology. 2010;49(3):433–40.CrossRefPubMed Nakashima R, Imura Y, Kobayashi S, Yukawa N, Yoshifuji H, Nojima T, et al. The RIG-I-like receptor IFIH1/MDA5 is a dermatomyositis-specific autoantigen identified by the anti-CADM-140 antibody. Rheumatology. 2010;49(3):433–40.CrossRefPubMed
22.
Zurück zum Zitat Frostegard J, Ulfgren AK, Nyberg P, Hedin U, Swedenborg J, Andersson U, et al. Cytokine expression in advanced human atherosclerotic plaques: dominance of pro-inflammatory (Th1) and macrophage-stimulating cytokines. Atherosclerosis. 1999;145(1):33–43.CrossRefPubMed Frostegard J, Ulfgren AK, Nyberg P, Hedin U, Swedenborg J, Andersson U, et al. Cytokine expression in advanced human atherosclerotic plaques: dominance of pro-inflammatory (Th1) and macrophage-stimulating cytokines. Atherosclerosis. 1999;145(1):33–43.CrossRefPubMed
23.
Zurück zum Zitat Brooke MH, Kaiser KK. Three “myosin adenosine triphosphatase” systems: the nature of their pH lability and sulfhydryl dependence. J Histochem Cytochem. 1970;18(9):670–2.CrossRefPubMed Brooke MH, Kaiser KK. Three “myosin adenosine triphosphatase” systems: the nature of their pH lability and sulfhydryl dependence. J Histochem Cytochem. 1970;18(9):670–2.CrossRefPubMed
24.
Zurück zum Zitat Scott W, Stevens J. Binder–Macleod SA. Human skeletal muscle fiber type classifications. Phys Ther. 2001;81(11):1810–6.PubMed Scott W, Stevens J. Binder–Macleod SA. Human skeletal muscle fiber type classifications. Phys Ther. 2001;81(11):1810–6.PubMed
25.
Zurück zum Zitat Chen Y-W, Zhao P, Borup R, Hoffman EP. Expression profiling in the muscular dystrophies identification of novel aspects of molecular pathophysiology. J Cell Biol. 2000;151(6):1321–36.CrossRefPubMedPubMedCentral Chen Y-W, Zhao P, Borup R, Hoffman EP. Expression profiling in the muscular dystrophies identification of novel aspects of molecular pathophysiology. J Cell Biol. 2000;151(6):1321–36.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Cooper ST, Lo HP, North KN. Single section Western blot: improving the molecular diagnosis of the muscular dystrophies. Neurology. 2003;61(1):93–7.CrossRefPubMed Cooper ST, Lo HP, North KN. Single section Western blot: improving the molecular diagnosis of the muscular dystrophies. Neurology. 2003;61(1):93–7.CrossRefPubMed
27.
Zurück zum Zitat Bruce CR, Risis S, Babb JR, Yang C, Kowalski GM, Selathurai A, et al. Overexpression of sphingosine kinase 1 prevents ceramide accumulation and ameliorates muscle insulin resistance in high-fat diet-fed mice. Diabetes. 2012;61(12):3148–55.CrossRefPubMedPubMedCentral Bruce CR, Risis S, Babb JR, Yang C, Kowalski GM, Selathurai A, et al. Overexpression of sphingosine kinase 1 prevents ceramide accumulation and ameliorates muscle insulin resistance in high-fat diet-fed mice. Diabetes. 2012;61(12):3148–55.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Higgs BW, Liu Z, White B, Zhu W, White WI, Morehouse C, et al. Patients with systemic lupus erythematosus, myositis, rheumatoid arthritis and scleroderma share activation of a common type I interferon pathway. Ann Rheum Dis. 2011;70(11):2029–36.CrossRefPubMed Higgs BW, Liu Z, White B, Zhu W, White WI, Morehouse C, et al. Patients with systemic lupus erythematosus, myositis, rheumatoid arthritis and scleroderma share activation of a common type I interferon pathway. Ann Rheum Dis. 2011;70(11):2029–36.CrossRefPubMed
29.
Zurück zum Zitat de Jong TD, Vosslamber S, Mantel E, de Ridder S, Wesseling JG, van der Pouw Kraan TC, et al. Physiological evidence for diversification of IFNα-and IFNβ-mediated response programs in different autoimmune diseases. Arthritis Res Ther. 2016;18(1):1.CrossRef de Jong TD, Vosslamber S, Mantel E, de Ridder S, Wesseling JG, van der Pouw Kraan TC, et al. Physiological evidence for diversification of IFNα-and IFNβ-mediated response programs in different autoimmune diseases. Arthritis Res Ther. 2016;18(1):1.CrossRef
30.
Zurück zum Zitat Cappelletti C, Baggi F, Zolezzi F, Biancolini D, Beretta O, Severa M, et al. Type I interferon and Toll-like receptor expression characterizes inflammatory myopathies. Neurology. 2011;76(24):2079–88.CrossRefPubMed Cappelletti C, Baggi F, Zolezzi F, Biancolini D, Beretta O, Severa M, et al. Type I interferon and Toll-like receptor expression characterizes inflammatory myopathies. Neurology. 2011;76(24):2079–88.CrossRefPubMed
31.
Zurück zum Zitat Zhou X, Dimachkie MM, Xiong M, Tan FK, Arnett FC. cDNA microarrays reveal distinct gene expression clusters in idiopathic inflammatory myopathies. Med Sci Monit. 2004;10(7):BR191–7.PubMed Zhou X, Dimachkie MM, Xiong M, Tan FK, Arnett FC. cDNA microarrays reveal distinct gene expression clusters in idiopathic inflammatory myopathies. Med Sci Monit. 2004;10(7):BR191–7.PubMed
32.
Zurück zum Zitat Walsh RJ, Kong SW, Yao Y, Jallal B, Kiener PA, Pinkus JL, et al. Type I interferon-inducible gene expression in blood is present and reflects disease activity in dermatomyositis and polymyositis. Arthritis Rheum. 2007;56(11):3784–92.CrossRefPubMedPubMedCentral Walsh RJ, Kong SW, Yao Y, Jallal B, Kiener PA, Pinkus JL, et al. Type I interferon-inducible gene expression in blood is present and reflects disease activity in dermatomyositis and polymyositis. Arthritis Rheum. 2007;56(11):3784–92.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Greenberg S, Higgs B, Morehouse C, Walsh R, Kong SW, Brohawn P, Zhu W, Amato A, Salajegheh M, White B. Relationship between disease activity and type 1 interferon-and other cytokine-inducible gene expression in blood in dermatomyositis and polymyositis. Genes Immun. 2012;13(3):207–213. Greenberg S, Higgs B, Morehouse C, Walsh R, Kong SW, Brohawn P, Zhu W, Amato A, Salajegheh M, White B. Relationship between disease activity and type 1 interferon-and other cytokine-inducible gene expression in blood in dermatomyositis and polymyositis. Genes Immun. 2012;13(3):207–213.
34.
Zurück zum Zitat Hawke TJ, Kanatous SB, Martin CM, Goetsch SC, Garry DJ. Rad is temporally regulated within myogenic progenitor cells during skeletal muscle regeneration. Am J Physiol Cell Physiol. 2006;290(2):C379–87.CrossRefPubMed Hawke TJ, Kanatous SB, Martin CM, Goetsch SC, Garry DJ. Rad is temporally regulated within myogenic progenitor cells during skeletal muscle regeneration. Am J Physiol Cell Physiol. 2006;290(2):C379–87.CrossRefPubMed
35.
Zurück zum Zitat Binder EB. The role of FKBP5, a co-chaperone of the glucocorticoid receptor in the pathogenesis and therapy of affective and anxiety disorders. Psychoneuroendocrinology. 2009;34:S186–95.CrossRefPubMed Binder EB. The role of FKBP5, a co-chaperone of the glucocorticoid receptor in the pathogenesis and therapy of affective and anxiety disorders. Psychoneuroendocrinology. 2009;34:S186–95.CrossRefPubMed
36.
Zurück zum Zitat Loell I, Helmers S, Dastmalchi M, Alexanderson H, Munters L, Nennesmo I, et al. Higher proportion of fast‐twitch (type II) muscle fibres in idiopathic inflammatory myopathies—evident in chronic but not in untreated newly diagnosed patients. Clin Physiol Funct Imaging. 2011;31(1):18–25.CrossRefPubMed Loell I, Helmers S, Dastmalchi M, Alexanderson H, Munters L, Nennesmo I, et al. Higher proportion of fast‐twitch (type II) muscle fibres in idiopathic inflammatory myopathies—evident in chronic but not in untreated newly diagnosed patients. Clin Physiol Funct Imaging. 2011;31(1):18–25.CrossRefPubMed
37.
Zurück zum Zitat Dastmalchi M, Alexanderson H, Loell I, Stahlberg M, Borg K, Lundberg IE, et al. Effect of physical training on the proportion of slow-twitch type I muscle fibers, a novel nonimmune-mediated mechanism for muscle impairment in polymyositis or dermatomyositis. Arthritis Rheum. 2007;57(7):1303–10.CrossRefPubMed Dastmalchi M, Alexanderson H, Loell I, Stahlberg M, Borg K, Lundberg IE, et al. Effect of physical training on the proportion of slow-twitch type I muscle fibers, a novel nonimmune-mediated mechanism for muscle impairment in polymyositis or dermatomyositis. Arthritis Rheum. 2007;57(7):1303–10.CrossRefPubMed
38.
40.
Zurück zum Zitat Nikolova-Karakashian MN, Reid MB. Sphingolipid metabolism, oxidant signaling, and contractile function of skeletal muscle. Antioxid Redox Signal. 2011;15(9):2501–17.CrossRefPubMedPubMedCentral Nikolova-Karakashian MN, Reid MB. Sphingolipid metabolism, oxidant signaling, and contractile function of skeletal muscle. Antioxid Redox Signal. 2011;15(9):2501–17.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Ferreira LF, Moylan JS, Stasko S, Smith JD, Campbell KS, Reid MB. Sphingomyelinase depresses force and calcium sensitivity of the contractile apparatus in mouse diaphragm muscle fibers. J Appl Physiol. 2012;112(9):1538–45.CrossRefPubMedPubMedCentral Ferreira LF, Moylan JS, Stasko S, Smith JD, Campbell KS, Reid MB. Sphingomyelinase depresses force and calcium sensitivity of the contractile apparatus in mouse diaphragm muscle fibers. J Appl Physiol. 2012;112(9):1538–45.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Bingham A, Mamyrova G, Rother KI, Oral E, Cochran E, Premkumar A, et al. Predictors of acquired lipodystrophy in juvenile-onset dermatomyositis and a gradient of severity. Medicine. 2008;87(2):70.CrossRefPubMedPubMedCentral Bingham A, Mamyrova G, Rother KI, Oral E, Cochran E, Premkumar A, et al. Predictors of acquired lipodystrophy in juvenile-onset dermatomyositis and a gradient of severity. Medicine. 2008;87(2):70.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Lee LA, Hobbs KF. Lipodystrophy and metabolic abnormalities in a case of adult dermatomyositis. J Am Acad Dermatol. 2007;57(5):S85–7.CrossRefPubMed Lee LA, Hobbs KF. Lipodystrophy and metabolic abnormalities in a case of adult dermatomyositis. J Am Acad Dermatol. 2007;57(5):S85–7.CrossRefPubMed
44.
Zurück zum Zitat Munters LA, Loell I, Ossipova E, Raouf J, Dastmalchi M, Lindroos E, Chen YW, Esbjörnsson M, Korotkova M, Alexanderson H. Endurance Exercise Improves Molecular Pathways of Aerobic Metabolism in Patients with Myositis. Arthritis and Rheumatology; 2016. doi:10.1002/art.39624. Munters LA, Loell I, Ossipova E, Raouf J, Dastmalchi M, Lindroos E, Chen YW, Esbjörnsson M, Korotkova M, Alexanderson H. Endurance Exercise Improves Molecular Pathways of Aerobic Metabolism in Patients with Myositis. Arthritis and Rheumatology; 2016. doi:10.​1002/​art.​39624.
Metadaten
Titel
Effects on muscle tissue remodeling and lipid metabolism in muscle tissue from adult patients with polymyositis or dermatomyositis treated with immunosuppressive agents
verfasst von
Ingela Loell
Joan Raouf
Yi-Wen Chen
Rongye Shi
Inger Nennesmo
Helene Alexanderson
Maryam Dastmalchi
Kanneboyina Nagaraju
Marina Korotkova
Ingrid E. Lundberg
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Arthritis Research & Therapy / Ausgabe 1/2016
Elektronische ISSN: 1478-6362
DOI
https://doi.org/10.1186/s13075-016-1033-y

Weitere Artikel der Ausgabe 1/2016

Arthritis Research & Therapy 1/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

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