Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2012

Open Access 01.12.2012 | Research

Cellular sources of dysregulated cytokines in relapsing-remitting multiple sclerosis

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2012

Abstract

Background

Numerous cytokines are implicated in the immunopathogenesis of multiple sclerosis (MS), but studies are often limited to whole blood (WB) or peripheral blood mononuclear cells (PBMCs), thereby omitting important information about the cellular origin of the cytokines. Knowledge about the relation between blood and cerebrospinal fluid (CSF) cell expression of cytokines and the cellular source of CSF cytokines is even more scarce.

Methods

We studied gene expression of a broad panel of cytokines in WB from relapsing-remitting multiple sclerosis (RRMS) patients in remission and healthy controls (HCs). Subsequently we determined the gene expression of the dysregulated cytokines in isolated PBMC subsets (CD4+, CD8+T-cells, NK-cells, B-cells, monocytes and dendritic cells) from RRMS patients and HCs and in CSF-cells from RRMS patients in clinical relapse and non-inflammatory neurological controls (NIND).

Results

RRMS patients had increased expression of IFN-gamma (IFNG), interleukin (IL) 1-beta (IL1B), IL7, IL10, IL12A, IL15, IL23, IL27, lymphotoxin-alpha (LTA) and lymphotoxin-beta (LTB) in WB. In PBMC subsets the main sources of pro-inflammatory cytokines were T- and B-cells, whereas monocytes were the most prominent source of immunoregulatory cytokines. In CSF-cells, RRMS patients had increased expression of IFNG and CD19 and decreased expression of IL10 and CD14 compared to NINDs. CD19 expression correlated with expression of IFNG, IL7, IL12A, IL15 and LTA whereas CD14 expression correlated with IL10 expression.

Conclusions

Using a systematic approach, we show that expression of pro-inflammatory cytokines in peripheral blood primarily originates from T- and B-cells, with an important exception of IFNG which is most strongly expressed by NK-cells. In CSF-cell studies, B-cells appear to be enriched in RRMS and associated with expression of pro-inflammatory cytokines; contrarily, monocytes are relatively scarce in CSF from RRMS patients and are associated with IL10 expression. Thus, our findings suggest a pathogenetic role of B-cells and an immunoregulatory role of monocytes in RRMS.
Hinweise

Competing interests

JRC received honoraria for lecturing from Biogen-Idec. DH has received funding for travel from Biogen Idec, Merck-Serono and Sanofi-Aventis; and received speaker honoraria from Sanofi-Aventis and Biogen Idec. LB, MK and HBS report no disclosures. PSS has served on scientific advisory boards for Biogen Idec, Merck Serono, Novartis, Genmab, TEVA, Elan and GSK; and has been on steering committees or independent data monitoring boards in clinical trials sponsored by Merck Serono, Genmab, TEVA, GSK and Bayer Schering. He has received funding of travel for these activities, has served as Editor-in-Chief of the European Journal of Neurology, and is an editorial board member for Therapeutic Advances in Neurological Disorders and Multiple Sclerosis.He has received speaker honoraria from Biogen Idec, Merck Serono, TEVA, Bayer Schering, Sanofi-aventis, and Novartis. His department has received research support from Biogen Idec, Bayer Schering, Merck Serono, TEVA, Baxter, Sanofi-Aventis, BioMS, Novartis, Bayer, RoFAR, Roche, Genzyme, and from the Danish Multiple Sclerosis Society, the Danish Medical Research Council, and the European Union Sixth Framework Programme: Life Sciences, Genomics and Biotechnology for Health. FS has served on scientific advisory boards for and received funding for travel from Biogen Idec, Merck-Serono, Novartis, Sanofi-Aventis and Teva; and has served as a consultant for Biogen Idec and Novo Nordisk; received speaker honoraria from Bayer-Schering, Biogen Idec, Merck-Serono, Novartis, Sanofi-Aventis and Schering-Ploug. He has received research support from Biogen Idec, Merck-Serono, Novartis and Sanofi-Aventis; and serves as section editor on Multiple Sclerosis and Related Disorders.

Authors’ contributions

JRC, LB, DH, MK, HBS and FS collected the samples, performed the laboratory analyses and analyzed data. JRC and FS wrote the first draft of the manuscript. All authors participated in the interpretation of the data and contributed to the critical review of the manuscript. All authors read and approved the final version of the manuscript.
Abkürzungen
APC
Antigen-presenting cell
CNS
Central nervous system
CSF
Cerebrospinal fluid
CT
Threshold cycle
DC
Dendritic cell
EAE
Experimental autoimmune encephalomyelitis
EDSS
Expanded Disability Status Scale
FDR
False discovery rate
HC
Healthy controls
IFN
Interferon
IFNG
Interferon-gamma
IL
Interleukin
LTA
Lymphotoxin-alpha
LTB
Lymphotoxin-beta
MS
Multiple sclerosis
NIND
Non-Inflammatory neurological disease
NK-cell
Natural killer cell
NR
Normalization ratio
PBMC
Peripheral blood mononuclear cell
RRMS
Relapsing-remitting MS
RT-PCR
Real-time polymerase chain reactions
Th1
T-helper type 1
Th17
T-helper type 17
WB
Whole blood.

Introduction

Multiple sclerosis (MS) is a chronic, immune-mediated disease of the central nervous system (CNS). An interplay among genetic susceptibility and environmental factors is implicated in the pathogenesis of MS. In relapsing-remitting MS (RRMS), disability develops with relapses. The pathological correlates of clinical relapses are focal, transient attacks of immune cells on myelin and axons, resulting in the formation of an MS plaque [1]. In pathology studies of brain tissue from RRMS patients, T- and B-cells are seen accumulating in perivascular cuffs near MS plaques and to a lesser extent in the parenchyma, while monocytes and macrophages are seen mostly in the lesion parenchyma. In the cerebrospinal fluid (CSF) many MS patients show a mild pleocytosis. T-cells are the major cell subset in CSF, and compared to non-inflammatory neurological disease (NIND) an increase in B-cells and plasma cells and a decrease in monocytes and natural killer (NK) cells is observed in MS patients [2]. The relation between the peripheral immune system and CNS inflammation in RRMS is underscored by the efficacy of treatment with the monoclonal antibodies natalizumab [3] and rituximab [4], which exert their effect on peripheral immune cells, but have a major impact on disease activity, cell numbers, levels of cytokines and markers of tissue damage in CSF [57]. During immune attacks on myelin, peripheral activation and subsequent migration of autoreactive T-helper type 1 (Th1) and Th17 cells to the CNS is an essential step [8]. How the T-cells become activated remains unclear, but antigen-presenting cells (APCs) located in peripheral lymph nodes [9] and the subarachnoidal space [10] are likely to be involved.
Gene expression studies of brain tissue, peripheral blood mononuclear cells (PBMCs) and whole blood (WB) from MS patients have shown dysregulated expression of cytokines, chemokines, and transcription factors related to immune activation [11]. Interferon (IFN)-gamma, the Th1 signature cytokine, has been implicated in the pathogenesis of MS, and induced disease activity in a clinical trial [12], although in the animal model experimental autoimmune encephalomyelitis (EAE), Ifng knockout mice developed severe disease [13]. In addition to Th1-cells, Th17-cells that secrete interleukin (IL)-17 are believed to be important in the pathogenesis [8].
Since gene expression studies of molecules used in biomarker studies are mostly carried out on WB or PBMCs, the cellular origin of cytokines is usually not determined. To date, no study has systematically analyzed the cellular origin of dysregulated cytokines in MS patients. Furthermore little is known about cytokine expression in CSF-cells and the cellular source of CSF cytokines. CSF studies are sparse due to the limited availability of CSF, but studies linking peripheral and CSF immune responses are central to understanding the immunopathogenesis of MS, and to translate the many findings in studies of peripheral immune activation.
In the present study, we used gene expression analysis to identify dysregulated cytokines in WB from RRMS patients in clinical remission and subsequently investigated the cellular source of these cytokines in isolated PBMC subsets. Furthermore, to relate the findings in peripheral blood to CNS inflammation, we studied gene expression of the dysregulated cytokines in CSF-cells from RRMS patients in relapse, and determined whether it correlated with markers for T-, B- and NK-cells and monocytes in the CSF.

Materials and methods

Subjects

An initial cohort of 39 untreated RRMS patients in clinical remission and 39 healthy controls (HCs) was recruited for studies of gene expression in WB (Table 1). A second cohort of four untreated RRMS patients in remission and four HCs was recruited for studies of gene expression in isolated PBMC subsets. Finally a third cohort of 17 untreated RRMS patients in relapse and 10 NIND patients (3 with spinal stenosis, 3 with herniated lumbar discs and 4 with low back pain) were recruited for studies on CSF-cells and PBMCs; CSF sampling was done prior to initiation of eventual steroid pulse therapy. All RRMS cohorts studied consisted of both recent onset RRMS and RRMS with longer disease duration (Table 1). The studies were approved by the local scientific ethics committee and informed consent was obtained from all patients and healthy controls.
Table 1
Demographic and clinical characteristics of the cohorts
 
RRMS patients
Control groups
 
Median age
Female%
Median EDSS
Duration
Mean age
Female%
Whole blood studies
34.0 (28.0 to 39.0)
59%
2.0 (1.0 to 3.0)
5.0 (2.0 to 7.0)
32.0 (29.0 to 38.0)
58%
Cell subset studies
38.0 (30.5 to 47.0)
75%
1.5 (1.0 to 3.1)
5.0 (1.3 to 12.5)
33.5 (24.5 to 36.5)
75%
CSF cell studies
37.5 (31.3 to 42.8)
59%
3.5 (3.0 to 6.4)
6.0 (1.5 to 9.75)
51.5 (45.0 to 59.0)
70%
Median values with interquartile ranges in brackets. EDSS, Expanded Disability Status Scale; RRMS, relapsing-remitting multiple sclerosis.

Gene expression studies

WB was sampled in PAXgene tubes and RNA extracted using PAXgene Blood RNA Kit (PreAnalytiX, Hombrechtikon, Switzerland); cDNA synthesis was done with High Capacity cDNA Reverse Transcription Kit (Applied Biosystems, Foster City, California, USA). For studies of gene expression in PBMC subsets, PBMCs were isolated using Lymphoprep (Axis-Shield, Oslo, Norway) and subsets of CD4+T-cells, CD8+T-cells, NK-cells, B-cells, monocytes and dendritic cells (DC) were isolated using MACS cell separation kits (CD4+T Cell Isolation Kit, CD8+T Cell Isolation Kit, NK Cell Isolation Kit, CD19 MicroBeads, CD14 MicroBeads and Blood Dendritic Cell Isolation Kit) and an autoMACS separator (all from Miltenyi Biotec, Bergisch Gladbach, Germany). Mean purities of the PBMC subsets were above 93%, except for NK-cells which had a mean purity of 73%. RNA was extracted from 80,000 to 200,000 cells of the obtained subsets with PicoPure RNA Isolation Kit (Arcturus, Mountain View, California, USA). cDNA synthesis was done with qScript cDNA SuperMix (Quanta BioSciences, Gaithersburg, Maryland, USA). For CSF-cell gene expression studies, CSF was obtained by lumbar puncture within 30 days from the onset of a relapse with no evidence of spontaneous recovery. Blood was sampled on the same day as the lumbar puncture and PBMCs were isolated with Lymphoprep. We snap froze at least 5,000 CSF-cells and 200,000 PBMCs for later RNA extraction with PicoPure RNA Isolation Kit and cDNA synthesis with High Capacity cDNA Reverse Transcription Kit.
In the selection of the genes of interest (Table 2), we focused on genes that have been shown to be involved in the pathogenesis of MS, genes involved in the expression of Th1 and Th17 cytokines, and genes expressed in APCs. In a pilot study in the WB cohort we excluded IL1A, IL2, IL4, IL5, IL6, IL9, IL12B, IL13, IL17A, IL17F, IL21, IL33, GM-CSF and BDNF from analysis as candidate biomarkers due to low expression levels in WB samples. Real-time polymerase chain reactions (RT-PCR) were performed with TaqMan Gene Expression Assays on a 7500 Real-Time PCR System (Applied Biosystems, USA). Threshold cycle (CT) values were calculated using SDS software (Applied Biosystems, USA). In CSF-cell samples without amplification the expression value was arbitrarily set to 0. The relative mRNA transcript expression was calculated by the comparative CT method also referred to as the 2-ΔΔCT method, using GAPDH as reference gene. Expression values were normalized to a HC PBMC cDNA pool resulting in a normalization ratio (NR):
https://static-content.springer.com/image/art%3A10.1186%2F1742-2094-9-215/MediaObjects/12974_2012_719_Equ1_HTML.gif
Table 2
List of TaqMan Gene Expression Assays used in the gene expression studies
Gene Symbol
Assay number
GenBank
Whole blood
PBMC populations
CSF cells
GAPDH
Hs99999905_m1
NM_002046
·
·
·
IFNG
Hs99999041_m1
NM_000619
·
·
·
IL1B
Hs00174097_m1
NM_000576
·
·
·
IL1RN
Hs00277299_m1
NM_000577
·
  
IL7
Hs00174202_m1
NM_000880
·
·
·
IL10
Hs00174086_m1
NM_000572
 
·
·
IL12A
Hs00168405_m1
NM_000882
·
·
·
IL15
Hs00542571_m1
NM_172175
·
·
·
IL18
Hs00155517_m1
NM_001562
·
  
IL23
Hs00372324_m1
NM_016584
 
·
·
IL27
Hs00377366_m1
NM_145659
·
·
·
EBI3
Hs00194957_m1
NM_005755
·
  
LTA
Hs00236874_m1
NM_000595
·
·
·
LTB
Hs00242739_m1
NM_009588
·
·
·
TGFB1
Hs99999918_m1
NM_000660
·
  
TNF
Hs00174128_m1
NM_000594
·
  
PRF1
Hs00169473_m1
NM_005041
·
  
CD3d
Hs00174158_m1
NM_000732
 
·
·
CD14
Hs00169122_g1
NM_000591
 
·
·
CD19
Hs01047407_m1
NM_001770
 
·
·
CD56
Hs00941824_m1
NM_181351
 
·
·
Assay numbers listed represents TaqMan Gene Expression Assays used. CSF, cerebrospinal fluid; GenBank, GenBank accession numbers; PBMC, peripheral blood mononuclear cell.

Statistical analysis

Statistical analysis was performed using PASW 18 software (IBM, Armonk, New York, USA). For comparison between independent groups, non-parametric Mann–Whitney tests were used. Wilcoxon signed rank tests were used for analysis of paired samples. For correlation analysis, we used Spearman's rank correlation coefficient. Changes in the expression of mRNA were considered significant for P <0.05. Since the analysis of gene expression data involved multiple comparisons, we also used the false discovery rate (FDR) method to calculate q-values [14]. Q-values were considered significant for q <0.05.

Results

Gene expression in whole blood from RRMS patients in remission

Initially, we compared the WB expression of cytokine genes in RRMS patients in remission and HCs. We found significantly increased expression of IFN-gamma (IFNG), IL1B, IL7 IL12A IL15 IL27, lymphotoxin-alpha (LTA) and lymphotoxin-beta (LTB) in RRMS (Figure 1). In previous studies conducted in the same cohort we found increased expression of IL10 and IL23[15].

Gene expression in PBMC cell subpopulations from RRMS patients in remission

To substantiate the findings of increased cytokine gene expression in WB from RRMS patients in remission, we conducted gene expression studies on PBMC subsets from four RRMS patients in remission and four HCs. Due to the low number of subjects, we did not expect to find differences between the two groups. As shown in Figure 2, the cellular sources of IFNG are mainly NK-cells, CD8+T-cells and, to a lesser extent, CD4+T-cells. This is not unexpected, but highlights NK-cells as a major source of IFNG and thus questions the use of IFNG as a specific Th1 biomarker in peripheral blood studies without detailing the cellular source. For IL1B, the cellular sources are mainly monocytes and DCs. Notably, IL1B expression in WB is higher than in PBMCs, reflecting that granulocytes are a major source of IL1B in WB. IL10 is expressed in monocytes, CD4+ and CD8+T-cells. IL7 and IL12A are expressed mainly in B-cells and IL15 mainly in B-cells and monocytes. For IL23, the cellular sources are CD4+T-cells, CD8+T-cells and B-cells. Unexpectedly, we observed low expression in monocytes and DCs. This finding contradicts the current understanding of IL23, which is thought to be an APC cytokine. However, since IL23A transcripts are translated into IL23p19 protein, which together with IL12p40 form bioactive IL23, and IL12p40 production is known to be limited to activated monocytes and DCs [16] and B-cells [17], the biological role of IL23 expression in T-cells is unclear. Thus, our findings indicate that in vivo WB IL23 gene expression is not suitable for studying IL23, which should instead be studied at the protein level as IL-23 p19/IL-12 p40 heterodimer. IL27 expression is restricted to monocytes. LTA is mainly expressed in CD4+T-cells, CD8+T-cells and B-cells and, to a lesser extent, in NK-cells while LTB is mainly expressed in B-cells, CD4+T-cells and at lower levels in CD8+T-cells.

Gene expression in PBMCs and CSF-cells from RRMS relapse patients

Next, we wanted to relate the findings of dysregulated cytokines in peripheral blood cells with CNS inflammation. We analyzed the cytokine expression in CSF-cells and PBMCs from a cohort of RRMS relapsed and NIND patients (Table 3). IL10 expression was significantly enriched in CSF-cells compared to PBMCs with a 9-fold increase in RRMS relapse patients and a 42-fold increase in NINDs. LTB expression was higher in CSF-cells than in PBMCs with a 2.4-fold increase in RRMS relapsed patients and a 1.7-fold increase in NINDs. IL1B expression in CSF-cells was 0.18-fold lower than in PBMCs in RRMS relapsed patients. Compared with NIND, RRMS patients had increased IFNG and decreased IL10 expression in CSF-cells. The IFNG and LTA CSF/PBMC ratios were significantly higher in RRMS relapsed patients, while the IL10 CSF/PBMC ratio was lower in RRMS than in NIND patients. In CSF-cells IL7 and IL15 expression was only detectable in a few samples (all from RRMS patients).
Table 3
Cytokine expression in cerebrospinal fluid cells and peripheral blood mononuclear cells
  
RRMS relapse
NIND
RRMS relapse vs NIND
 
Mean CT value
N
Median (IQR)
CSF/PBMC p-value
N
Median (IQR)
CSF/PBMC p-value
p-value
q-value
IFNG PBMC NR
32.3
15
4.10 (2.74 to 10.21)
 
10
5.25 (2.65 to 9.12)
 
0.78
0.37
IFNG CSF NR
34.9
15
7.22 (0.00 to 16.42)
 
10
0.00 (0.00 to 0.00)
 
0.004
0.02
IFNG CSF/PBMC Ratio
 
15
0.71 (0.00 to 2.31)
0.87
10
0.00 (0.00 to 0.00)
 
0.004
0.02
IL1B PBMC NR
32.4
16
1.13 (0.88 to 2.14)
 
10
1.81 (1.05 to 2.70)
 
0.19
0.15
IL1B CSF NR
36.7
14
0.21 (0.00 to 1.09)
 
10
2.70 (0.00 to 6.94)
 
0.36
0.23
IL1B CSF/PBMC Ratio
 
13
0.18 (0.00 to 0.92)
0.02
10
1.96 (0.00 to 6.40)
0.29
0.42
0.24
IL7 PBMC NR
34.0
16
0.41 (0.21 to 1.17)
 
10
0.31 (0.18 to 0.46)
 
0.40
0.24
IL7 CSF NR
36.1
13
0.00 (0.00 to 0.86)
 
10
0.00 (0.00 to 0.00)
 
0.06
0.09
IL7 CSF/PBMC Ratio
 
12
0.00 (0.00 to 2.51)
 
8
0.00 (0.00-0.00)
 
0.08
0.09
IL10 PBMC NR
34.3
16
2.75 (2.02 to 3.90)
 
10
2.95 (2.38 to 4.26)
 
0.54
0.27
IL10 CSF NR
35.3
17
23.51 (3.22 to 46.39)
 
10
116.78 (72.22 to 156.28)
 
0.001
0.01
IL10 CSF/PBMC Ratio
 
16
8.57 (0.79 to 12.71)
0.003
10
42.21 (24.04 to 66.18)
0.01
0.002
0.01
IL12A PBMC NR
37.5
16
0.49 (0.30 to 0.66)
 
10
0.54 (0.42 to 0.82)
 
0.53
0.27
IL12A CSF NR
38.3
15
0.00 (0.00 to 0.00)
 
10
0.00 (0.00 to 0.00)
 
0.24
0.17
IL12A CSF/PBMC Ratio
 
12
0.00 (0.00 to 0.00)
 
9
0.00 (0.00 to 0.00)
 
0.21
0.16
IL15 PBMC NR
36.3
16
1.19 (0.91 to 1.43)
 
10
1.10 (0.57 to 1.60)
 
0.79
0.37
IL15 CSF NR
37.3
14
0.00 (0.00 to 1.10)
 
10
0.00 (0.00 to 0.00)
 
0.07
0.09
IL15 CSF/PBMC Ratio
 
13
0.00 (0.00 to 0.97)
 
10
0.00 (0.00 to 0.00)
 
0.06
0.09
IL23 PBMC NR
34.6
16
0.80 (0.65 to 1.18)
 
10
0.80 (0.62 to 1.15)
 
1.00
0.44
IL23 CSF NR
36.8
14
1.12 (0.00 to 1.98)
 
10
0.00 (0.00 to 0.48)
 
0.10
0.10
IL23 CSF/PBMC Ratio
 
13
1.54 (0.00 to 3.17)
0.28
10
0.00 (0.00 to 1.50)
 
0.09
0.10
IL27 PBMC NR
35.1
16
4.74 (2.49 to 7.60)
 
10
8.36 (6.17 to 12.70)
 
0.045
0.09
IL27 CSF NR
ND
14
0.00 (0.00 to 0.00)
 
10
0.00 (0.00 to 0.00)
 
1.00
0.44
IL27 CSF/PBMC Ratio
 
12
0.00 (0.00 to 0.00)
 
10
0.00 (0.00 to 0.00)
 
1.00
0.44
LTA PBMC NR
34.3
16
1.74 (1.43 to 2.30)
 
10
2.11 (1.59 to 2.73)
 
0.37
0.23
LTA CSF NR
36.7
14
1.41 (0.00 to 3.20)
 
10
0.00 (0.00 to 0.00)
 
0.05
0.09
LTA CSF/PBMC Ratio
 
13
1.08 (0.00 to 1.82)
0.92
10
0.00 (0.00 to 0.00)
 
0.04
0.09
LTB PBMC NR
28.2
16
4.09 (2.44 to 4.99)
 
10
3.71 (3.06 to 5.54)
 
0.79
0.37
LTB CSF NR
33.5
13
8.05 (6.62 to 13.37)
 
10
8.82 (3.30 to 12.27)
 
0.42
0.23
LTB CSF/PBMC Ratio
 
12
2.37 (1.76 to 3.12)
0.002
10
1.70 (0.97 to 3.12)
0.04
0.13
0.11
Gene expression data are shown as the normalization ratio (NR). Mean cycle threshold (CT) is shown for each target. CSF/PBMC ratio represent fold change in expression between CSF cells and PBMCs. CSF/PBMC P-values represents Wilcoxon signed rank test results, and tests were only conducted if median CSF NR was >0. All other P-values represent Mann–Whitney tests for differences between groups, and q-values represent false discovery rate corrected P-values. Significant test-values are in bold. CSF, cerebrospinal fluid; IQR, Interquartile range; NIND, non-inflammatory neurological disease; PBMC; peripheral blood mononuclear cell; RRMS relapse, relapsing-remitting multiple sclerosis patients in relapse.

CSF-cell subsets and cytokine gene expression in RRMS relapse patients

To elucidate whether differences in cytokine gene expression are associated with the cell subsets present in CSF, we analysed gene expression of CD3d, CD14, CD19 and CD56 (markers of T-cells, monocytes, B-cells and NK-cells and NK T-cells, respectively) in CSF-cells (Table 4). Comparing expression of cell type markers in CSF-cells to PBMCs, we found significantly increased expression of CD3d in CSF-cells in RRMS relapse patients and increased CD14 expression in CSF-cells from NINDs. RRMS relapse patients had a decrease in the expression of CD14 and the CD14 CSF/PBMC ratio and an increase in expression of CD19 and the CD19 CSF/PBMC ratio compared with NIND patients.
Table 4
Expression of markers of cell types in cerebrospinal fluid cells and peripheral blood mononuclear cells
  
RRMS relapse
NIND
RRMS relapse vs NIND
 
Mean CT
N
Median (IQR)
CSF/PBMC p-value
N
Median (IQR)
CSF/PBMC p-value
p-value
q-value
CD3d PBMC NR
31.1
15
0.06 (0.04 to 0.08)
 
10
0.05 (0.04 to 0.07)
 
0.47
0.26
CD3d CSF NR
35.6
13
0.10 (0.08 to 0.23)
 
10
0.06 (0.02 to 0.15)
 
0.07
0.09
CD3d CSF/PBMC Ratio
 
11
2.37 (1.30 to 3.67)
0.01
10
1.12 (0.42 to 3.07)
0.39
0.23
0.17
CD14 PBMC NR
30.1
15
0.33 (0.27 to 0.38)
 
10
0.33 (0.29 to 0.49)
 
0.38
0.23
CD14 CSF NR
34.9
13
0.38 (0.07 to 0.52)
 
10
1.52 (0.94 to 2.40)
 
0.0004
0.01
CD14 CSF/PBMC Ratio
 
11
1.39 (0.30 to 2.15)
0.42
10
4.01 (3.54 to 5.16)
0.01
0.003
0.02
CD19 PBMC NR
36.1
15
0.14 (0.09 to 0.23)
 
10
0.08 (0.05 to 0.16)
 
0.05
0.09
CD19 CSF NR
37.1
13
0.00 (0.00 to 0.35)
 
10
0.00 (0.00 to 0.00)
 
0.03
0.09
CD19 CSF/PBMC Ratio
 
11
0.00 (0.00 to 0.89)
 
10
0.00 (0.00 to 0.00)
 
0.04
0.09
CD56 PBMC NR
34.9
15
0.15 (0.09 to 0.23)
 
10
0.16 (0.07 to 0.32)
 
0.96
0.43
CD56 CSF NR
37.8
13
0.00 (0.00 to 0.03)
 
10
0.00 (0.00 to 0.00)
 
0.11
0.10
CD56 CSF/PBMC Ratio
 
11
0.00 (0.00 to 0.00)
 
10
0.00 (0.00 to 0.00)
 
0.17
0.14
Gene expression data are shown as normalization ratio (NR). Mean cycle threshold (CT) is shown for each target. CSF/PBMC ratio represent fold change in expression between CSF cells and PBMCs. CSF/PBMC p-values represents Wilcoxon signed rank test results, and tests were only conducted if median CSF NR was >0. All other p-values represent Mann–Whitney tests for difference between groups, and q-values represent false discovery rate corrected p-values. Significant test-values are in bold. RRMS relapse: Relapsing-remitting multiple sclerosis patients in relapse. NIND: Non-inflammatory neurological disease. PBMC: Peripheral blood mononuclear cell. CSF: Cerebrospinal fluid. IQR: Interquartile range.
Correlations between CSF cytokine expression and cell type markers were analyzed using Spearman’s rank correlation analysis. Since several of the gene targets were only expressed in some patients, resulting in low statistical power, we analyzed correlations for the whole cohort and for the cohort of RRMS patients in relapse (Table 5). For the whole cohort, INFG and CD19 expression correlated positively, and INFG and CD14 expression correlated negatively. IL10 expression correlated positively with CD14 and negatively with CD19. IL10 expression in CSF-cells correlated significantly with CD14 expression, suggesting that monocytes are an important source of IL10 in CSF-cells. Consistent with the PBMC subset findings, we further found positive correlations between CD19 and IFNG, IL7, IL12A, IL15 and LTA; CD3d and LTB; CD56 and IL12A. These findings also reached statistical significance when FDR-corrected q-values were calculated. For RRMS relapse patients some of the correlations were confirmed, but none of them reached statistical significance when q-values were calculated.
Table 5
Correlations between cytokine and cell type marker expression in cerebrospinal fluid cells
   
RRMS relapse and NIND
RRMS relapse
CSF NR
N
R
p-value
q-value
N
R
p-value
q-value
IFNG
vs
CD3d
22
0.383
0.08
0.06
12
0.441
0.151
0.39
IFNG
vs
CD14
22
−0.606
0.003
0.008
12
0.018
0.956
0.90
IFNG
vs
CD19
22
0.538
0.010
0.02
12
0.111
0.731
1.04
IFNG
vs
CD56
22
0.372
0.09
0.06
12
0.019
0.954
0.93
IL1B
vs
CD3d
23
−0.453
0.03
0.03
13
0.064
0.837
0.89
IL1B
vs
CD14
23
0.126
0.57
0.27
13
0.067
0.829
0.91
IL1B
vs
CD19
23
0.168
0.44
0.21
13
0.435
0.137
0.39
IL1B
vs
CD56
23
0.110
0.62
0.28
13
0.285
0.345
0.73
IL7
vs
CD3d
23
0.090
0.68
0.30
13
−0.121
0.694
1.03
IL7
vs
CD14
23
−0.169
0.44
0.22
13
0.087
0.777
0.98
IL7
vs
CD19
23
0.600
0.002
0.009
13
0.513
0.073
0.36
IL7
vs
CD56
23
0.412
0.05
0.04
13
0.241
0.429
0.86
IL10
vs
CD3d
23
−0.506
0.01
0.02
13
−0.589
0.034
0.23
IL10
vs
CD14
23
0.773
0.00002
0.00011
13
0.541
0.056
0.32
IL10
vs
CD19
23
−0.438
0.04
0.04
13
−0.151
0.623
1.01
IL10
vs
CD56
23
−0.242
0.27
0.15
13
0.086
0.781
0.95
IL12A
vs
CD3d
23
0.075
0.73
0.31
13
−0.096
0.755
1.03
IL12A
vs
CD14
23
−0.327
0.13
0.08
13
−0.236
0.437
0.78
IL12A
vs
CD19
23
0.577
0.004
0.008
13
0.508
0.076
0.32
IL12A
vs
CD56
23
0.756
0.00003
0.00014
13
0.697
0.008
0.09
IL15
vs
CD3d
23
0.226
0.30
0.16
13
0.067
0.828
0.94
IL15
vs
CD14
23
−0.519
0.01
0.02
13
−0.450
0.123
0.42
IL15
vs
CD19
23
0.847
0.0000003
0.0000048
13
0.758
0.003
0.09
IL15
vs
CD56
23
0.469
0.02
0.03
13
0.349
0.242
0.59
IL23
vs
CD3d
23
0.004
0.98
0.39
13
0.034
0.912
0.91
IL23
vs
CD14
23
−0.396
0.06
0.05
13
−0.040
0.898
0.92
IL23
vs
CD19
23
0.340
0.11
0.07
13
0.177
0.562
0.96
IL23
vs
CD56
23
0.234
0.28
0.15
13
0.092
0.766
1.00
LTA
vs
CD3d
23
0.002
0.99
0.39
13
−0.121
0.693
1.07
LTA
vs
CD14
23
−0.420
0.05
0.04
13
−0.078
0.800
0.94
LTA
vs
CD19
23
0.563
0.005
0.009
13
0.461
0.113
0.43
LTA
vs
CD56
23
0.384
0.07
0.05
13
0.238
0.433
0.82
LTB
vs
CD3d
23
0.579
0.004
0.009
13
0.440
0.133
0.41
LTB
vs
CD14
23
−0.057
0.80
0.33
13
0.305
0.310
0.70
LTB
vs
CD19
23
−0.337
0.12
0.07
13
−0.702
0.008
0.13
LTB
vs
CD56
23
−0.304
0.16
0.09
13
−0.598
0.031
0.26
Gene expression data are shown as the normalization ratio (NR). Correlation analysis was done with Spearman’s rank correlation coefficient, and q-values represent the false discovery rate corrected P-values. Significant values are in bold. RRMS relapse: Relapsing-remitting multiple sclerosis patients in relapse. CSF cerebrospinal fluid; NIND, Non-inflammatory neurological disease; PBMC, Peripheral blood mononuclear cell.

Discussion

In the present study we find an increased expression of pro-inflammatory (INFG, IL1B, IL7, IL12A, IL15, IL23, LTA and LTB) and immunoregulatory cytokines (IL10 and IL27) in WB from RRMS patients in remission. In subsequent gene expression studies on PBMC subsets, our findings confirm the expected sources for many cytokines, but for others we reveal unexpected cell-types to be major sources. In general, T- and B-cells are the most frequent sources of the pro-inflammatory cytokines, and monocytes are the predominant source of the immunoregulatory cytokines. In CSF-cell studies, we demonstrate that the majority of the cytokines are expressed by CSF-cells, and that CSF-cells from RRMS patients have increased expression of IFNG and markers of T- and B-cells, whereas IL10 and the marker for monocytes have decreased expression. Finally, the B-cell marker CD19 correlated positively with many pro-inflammatory cytokines in CSF-cells.
We present a systematic approach to determine the cellular source of dysregulated cytokines in RRMS, which is critical in the interpretation of findings in cytokine biology. Our studies addressed the expression of genes that may serve as biomarkers of fundamental pathogenic processes in MS in fresh blood samples, which allows studying cytokine biology close to in vivo conditions, without the need for in vitro stimulation. However, in vivo studies have limitations, since they do not address the full potential for activation of the cells in the same way that in vitro studies do. Furthermore, gene expression does not necessarily result in translation into bioactive protein, and post-translational modifications or regulated secretion are not reflected by mRNA expression studies. The studies on isolated cell-subsets were generally performed on samples with high purity, but for NK-cells the purity was 73% on average. This could represent a potential bias, as genes not normally detected in NK-cells possibly will be detected. Thus, for genes with low expression levels in NK-cells, conclusions must be very cautious. On the other hand, the very high expression of IFNG by NK-cells is not likely to be a result of this impurity. The CSF-cell study had limited statistical power due to the relatively low number of subjects and the low amount of mRNA that could be extracted from these cells. This implies that more target molecules might have been detected if more mRNA had been available. Caution is therefore important in the interpretation of differences in expression levels between PBMCs and CSF-cells in MS patients and controls. Not only will differences in the cellular composition of CSF samples in MS patients and neurological controls confound the results obtained, but differences in the composition of blood and CSF-cell populations common to patients and controls should also be taken into account. Adding to this, specific cell-subsets might change their phenotype or gene expression patterns upon crossing the blood–brain-barrier [18, 19].
Looking into the possible roles of specific cytokines found to be dysregulated in our patients with RRMS, IFN-gamma is known to be present in brain lesions in MS patients [20], and IFN-gamma secretion is increased in T-cells from RRMS patients in the blood and even more in CSF. Experimental studies indicate that IFN-gamma, at least when expressed by CD4+T-cells, has important effects along with IL17 in the development of RRMS [8]. Our findings of increased IFNG in WB and CSF-cells in RRMS patients are in accordance with previous studies, but the finding of NK-cells and CD8+T-cells as the major sources highlights that IFNG expression in WB is associated with other cellular responses than Th1. As NK-cells have been attributed an immunoregulatory role in MS, this could explain the known ambiguous effects of IFN-gamma in MS and animal models of MS, and our finding emphasizes that further clarification of the function of IFN-gamma in MS requires studies of NK-cells and CD8+T-cells. The finding of increased IFNG expression in CSF-cells is more likely attributed a pro-inflammatory role in T-cells, and interestingly correlates with B-cells.
We have previously demonstrated negative correlation between expression of the immunoregulatory cytokine IL10 and the number of active magnetic resonance imaging lesions [21], and IL10 has been suggested to have beneficial effects in MS in numerous studies [22, 23]. IL10 is present in perivascular macrophages in MS lesions [24] and is increased in CSF from MS patients [25]. In blood, studies of IL10 expression have been conflicting [2527] but most biomarker studies point to a slight increase in IL10 gene expression in MS. Accordingly, we find IL10 expression to be increased in WB from RRMS in remission compared to HCs. In CSF-cells we find a decrease in IL10 mRNA expression in RRMS relapse compared to NIND patients. This corresponds to a study showing that IL10-protein is decreased during relapses [28], whereas data from a study on a mixed MS group and a non-inflammatory control group [29], reported increased IL10 mRNA expression in in CSF-cells from MS. The latter study used a mixed MS group where the majority was not in relapse, and hence a likely explanation for the conflicting results is the differences in the specific patient groups studied. Finally, since IL10 expression was pronounced in monocytes in our PBMC subset study and correlated negatively with CD3d and CD19 and positively with CD14 in CSF-cells, our findings support that monocytes may have an immunoregulatory role in MS. However, the decreased CSF-cell IL10 expression in RRMS compared to NIND patients could also simply reflect a relatively decreased frequency of monocytes in MS patients [2] or a change in the monocyte phenotype, since monocytes transmigrating across the blood–brain barrier change their phenotype [19], and CSF monocytes have decreased CD14 expression [18, 30].
IL7 and IL15 are IL-2 family cytokines regulating survival and activation of lymphocytes, and are of interest in MS research since they signal through the IL2- and IL7-receptors, which have shown strong association with MS in genome-wide association studies [31]. Expression of these cytokines is increased in MS brain lesions [32, 33], in CSF [34, 35] and in blood from MS patients [36, 37]. The finding of increased WB expression is in accordance with previous studies on blood cells. Since IL7 and IL15 both contribute to increased T-cell survival and Th1 induction, our findings could represent a B-cell and monocyte-driven pro-inflammatory response in the peripheral immune compartment of MS patients. On the other hand IL15 is also essential for NK-cell survival and IL15-deficiency in mice results in worsening of EAE [38], why caution in the interpretation of the IL15 findings should be stressed.
The finding of IL23 expression being most pronounced in T-and B-cells, make clear that a systematic approach can be necessary to derive proper conclusions about the biology behind cytokine gene expression, and in this particular example, that the biological significance of in vivo IL23 expression is unclear and will demand studies of IL23 expression in T-cells.
Lymphotoxin is present in MS lesions [39] and expression is increased in blood and CSF in MS [40, 41]. Being important for the crosstalk between APCs and T-cells and for the development of ectopic follicle-like structures seen in MS and other autoimmune diseases [42, 43] our finding of increased lymphotoxin in WB and CSF-cells points to a probable pro-inflammatory function of this cytokine in MS, although studies proving that the increased mRNA expression is associated with increased bioactivity are needed to substantiate this hypothesis.

Conclusion

In conclusion, we have confirmed previous findings of dysregulated cytokines in WB in MS patients, particularly increased expression of pro-inflammatory cytokines. A detailed analysis of the cytokine expression in PBMC subsets confirmed the expected origin for some cytokines, while other cytokines also were expressed in unexpected subsets. Most pro-inflammatory cytokines were expressed by B-cells in blood and correlated with CD19 in CSF-cells. Monocytes were the predominant source of immunoregulatory cytokines, and IL10 was decreased in CSF-cells compared to NINDs, and correlated with CD14 in CSF-cells. These findings correspond to studies on CSF-cells in MS [44, 45] and a study showing that the most variable cell parameter in CSF is the B-cell/monocyte ratio, which also correlates with disease progression [2]. Thus our study supports a central role of B-cells in the pathogenesis of MS. Recently this has been highlighted by studies proving the benefit of treatment with B cell-depleting antibodies in MS [46].

Acknowledgements

This work was supported by the Danish Council for Independent Research (grant 271-06-0246), the Danish Council for Strategic Research (grant 2142-08-0039), the Danish MS Society, the Warwara Larsen Foundation, the Johnsen Foundation, Brdr. Rønje Holding, Jeppe Juel Memorial Legacy and research grants from Biogen Idec, Merck Serono and Novartis.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

JRC received honoraria for lecturing from Biogen-Idec. DH has received funding for travel from Biogen Idec, Merck-Serono and Sanofi-Aventis; and received speaker honoraria from Sanofi-Aventis and Biogen Idec. LB, MK and HBS report no disclosures. PSS has served on scientific advisory boards for Biogen Idec, Merck Serono, Novartis, Genmab, TEVA, Elan and GSK; and has been on steering committees or independent data monitoring boards in clinical trials sponsored by Merck Serono, Genmab, TEVA, GSK and Bayer Schering. He has received funding of travel for these activities, has served as Editor-in-Chief of the European Journal of Neurology, and is an editorial board member for Therapeutic Advances in Neurological Disorders and Multiple Sclerosis.He has received speaker honoraria from Biogen Idec, Merck Serono, TEVA, Bayer Schering, Sanofi-aventis, and Novartis. His department has received research support from Biogen Idec, Bayer Schering, Merck Serono, TEVA, Baxter, Sanofi-Aventis, BioMS, Novartis, Bayer, RoFAR, Roche, Genzyme, and from the Danish Multiple Sclerosis Society, the Danish Medical Research Council, and the European Union Sixth Framework Programme: Life Sciences, Genomics and Biotechnology for Health. FS has served on scientific advisory boards for and received funding for travel from Biogen Idec, Merck-Serono, Novartis, Sanofi-Aventis and Teva; and has served as a consultant for Biogen Idec and Novo Nordisk; received speaker honoraria from Bayer-Schering, Biogen Idec, Merck-Serono, Novartis, Sanofi-Aventis and Schering-Ploug. He has received research support from Biogen Idec, Merck-Serono, Novartis and Sanofi-Aventis; and serves as section editor on Multiple Sclerosis and Related Disorders.

Authors’ contributions

JRC, LB, DH, MK, HBS and FS collected the samples, performed the laboratory analyses and analyzed data. JRC and FS wrote the first draft of the manuscript. All authors participated in the interpretation of the data and contributed to the critical review of the manuscript. All authors read and approved the final version of the manuscript.
Literatur
2.
Zurück zum Zitat Cepok S, Jacobsen M, Schock S, Omer B, Jaekel S, Boddeker I, Oertel WH, Sommer N, Hemmer B: Patterns of cerebrospinal fluid pathology correlate with disease progression in multiple sclerosis. Brain 2001, 124:2169–2176.CrossRefPubMed Cepok S, Jacobsen M, Schock S, Omer B, Jaekel S, Boddeker I, Oertel WH, Sommer N, Hemmer B: Patterns of cerebrospinal fluid pathology correlate with disease progression in multiple sclerosis. Brain 2001, 124:2169–2176.CrossRefPubMed
3.
Zurück zum Zitat Polman CH, O'Connor PW, Havrdova E, Hutchinson M, Kappos L, Miller DH, Phillips JT, Lublin FD, Giovannoni G, Wajgt A, Toal M, Lynn F, Panzara MA, Sandrock AW, AFFIRM Investigators: A randomized, placebo-controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med 2006, 354:899–910.CrossRefPubMed Polman CH, O'Connor PW, Havrdova E, Hutchinson M, Kappos L, Miller DH, Phillips JT, Lublin FD, Giovannoni G, Wajgt A, Toal M, Lynn F, Panzara MA, Sandrock AW, AFFIRM Investigators: A randomized, placebo-controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med 2006, 354:899–910.CrossRefPubMed
4.
Zurück zum Zitat Hauser SL, Waubant E, Arnold DL, Vollmer T, Antel J, Fox RJ, Bar-Or A, Panzara M, Sarkar N, Agarwal S, Langer-Gould A, Smith CH, HERMES Trial Group: B-cell depletion with rituximab in relapsing-remitting multiple sclerosis. N Engl J Med 2008, 358:676–688.CrossRefPubMed Hauser SL, Waubant E, Arnold DL, Vollmer T, Antel J, Fox RJ, Bar-Or A, Panzara M, Sarkar N, Agarwal S, Langer-Gould A, Smith CH, HERMES Trial Group: B-cell depletion with rituximab in relapsing-remitting multiple sclerosis. N Engl J Med 2008, 358:676–688.CrossRefPubMed
5.
Zurück zum Zitat Khademi M, Bornsen L, Rafatnia F, Andersson M, Brundin L, Piehl F, Sellebjerg F, Olsson T: The effects of natalizumab on inflammatory mediators in multiple sclerosis: prospects for treatment-sensitive biomarkers. Eur J Neurol 2009, 16:528–536.CrossRefPubMed Khademi M, Bornsen L, Rafatnia F, Andersson M, Brundin L, Piehl F, Sellebjerg F, Olsson T: The effects of natalizumab on inflammatory mediators in multiple sclerosis: prospects for treatment-sensitive biomarkers. Eur J Neurol 2009, 16:528–536.CrossRefPubMed
6.
Zurück zum Zitat del Pilar Martin M, Cravens PD, Winger R, Frohman EM, Racke MK, Eagar TN, Zamvil SS, Weber MS, Hemmer B, Karandikar NJ, Kleinschmidt-DeMasters BK, Stüve O: Decrease in the numbers of dendritic cells and CD4+ T cells in cerebral perivascular spaces due to natalizumab. Arch Neurol 2008, 65:1596–1603.CrossRefPubMed del Pilar Martin M, Cravens PD, Winger R, Frohman EM, Racke MK, Eagar TN, Zamvil SS, Weber MS, Hemmer B, Karandikar NJ, Kleinschmidt-DeMasters BK, Stüve O: Decrease in the numbers of dendritic cells and CD4+ T cells in cerebral perivascular spaces due to natalizumab. Arch Neurol 2008, 65:1596–1603.CrossRefPubMed
7.
Zurück zum Zitat Cross AH, Stark JL, Lauber J, Ramsbottom MJ, Lyons JA: Rituximab reduces B cells and T cells in cerebrospinal fluid of multiple sclerosis patients. J Neuroimmunol 2006, 180:63–70.CrossRefPubMedPubMedCentral Cross AH, Stark JL, Lauber J, Ramsbottom MJ, Lyons JA: Rituximab reduces B cells and T cells in cerebrospinal fluid of multiple sclerosis patients. J Neuroimmunol 2006, 180:63–70.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Kebir H, Ifergan I, Alvarez JI, Bernard M, Poirier J, Arbour N, Duquette P, Prat A: Preferential recruitment of interferon-gamma-expressing TH17 cells in multiple sclerosis. Ann Neurol 2009, 66:390–402.CrossRefPubMed Kebir H, Ifergan I, Alvarez JI, Bernard M, Poirier J, Arbour N, Duquette P, Prat A: Preferential recruitment of interferon-gamma-expressing TH17 cells in multiple sclerosis. Ann Neurol 2009, 66:390–402.CrossRefPubMed
9.
Zurück zum Zitat Van-Zwam M, Huizinga R, Melief MJ, Wierenga-Wolf AF, Van-Meurs M, Voerman JS, Biber KP, Boddeke HW, Hopken UE, Meisel C, Meisel A, Bechmann I, Hintzen RQ, T-Hart BA, Amor S, Laman JD, Boven LA: Brain antigens in functionally distinct antigen-presenting cell populations in cervical lymph nodes in MS and EAE. J Mol Med (Berl) 2009, 87:273–286.CrossRef Van-Zwam M, Huizinga R, Melief MJ, Wierenga-Wolf AF, Van-Meurs M, Voerman JS, Biber KP, Boddeke HW, Hopken UE, Meisel C, Meisel A, Bechmann I, Hintzen RQ, T-Hart BA, Amor S, Laman JD, Boven LA: Brain antigens in functionally distinct antigen-presenting cell populations in cervical lymph nodes in MS and EAE. J Mol Med (Berl) 2009, 87:273–286.CrossRef
10.
Zurück zum Zitat Kivisakk P, Imitola J, Rasmussen S, Elyaman W, Zhu B, Ransohoff RM, Khoury SJ: Localizing central nervous system immune surveillance: meningeal antigen-presenting cells activate T cells during experimental autoimmune encephalomyelitis. Ann Neurol 2009, 65:457–469.CrossRefPubMedPubMedCentral Kivisakk P, Imitola J, Rasmussen S, Elyaman W, Zhu B, Ransohoff RM, Khoury SJ: Localizing central nervous system immune surveillance: meningeal antigen-presenting cells activate T cells during experimental autoimmune encephalomyelitis. Ann Neurol 2009, 65:457–469.CrossRefPubMedPubMedCentral
11.
12.
Zurück zum Zitat Panitch HS, Hirsch RL, Haley AS, Johnson KP: Exacerbations of multiple sclerosis in patients treated with gamma interferon. Lancet 1987, 1:893–895.CrossRefPubMed Panitch HS, Hirsch RL, Haley AS, Johnson KP: Exacerbations of multiple sclerosis in patients treated with gamma interferon. Lancet 1987, 1:893–895.CrossRefPubMed
13.
Zurück zum Zitat Krakowski M, Owens T: Interferon-gamma confers resistance to experimental allergic encephalomyelitis. Eur J Immunol 1996, 26:1641–1646.CrossRefPubMed Krakowski M, Owens T: Interferon-gamma confers resistance to experimental allergic encephalomyelitis. Eur J Immunol 1996, 26:1641–1646.CrossRefPubMed
15.
Zurück zum Zitat Krakauer M, Sorensen P, Khademi M, Olsson T, Sellebjerg F: Increased IL-10 mRNA and IL-23 mRNA expression in multiple sclerosis: interferon-beta treatment increases IL-10 mRNA expression while reducing IL-23 mRNA expression. Mult Scler 2008, 14:622–630.CrossRefPubMed Krakauer M, Sorensen P, Khademi M, Olsson T, Sellebjerg F: Increased IL-10 mRNA and IL-23 mRNA expression in multiple sclerosis: interferon-beta treatment increases IL-10 mRNA expression while reducing IL-23 mRNA expression. Mult Scler 2008, 14:622–630.CrossRefPubMed
16.
Zurück zum Zitat Brentano F, Ospelt C, Stanczyk J, Gay RE, Gay S, Kyburz D: Abundant expression of the interleukin (IL)23 subunit p19, but low levels of bioactive IL23 in the rheumatoid synovium: differential expression and Toll-like receptor-(TLR) dependent regulation of the IL23 subunits, p19 and p40, in rheumatoid arthritis. Ann Rheum Dis 2009, 68:143–150.CrossRefPubMed Brentano F, Ospelt C, Stanczyk J, Gay RE, Gay S, Kyburz D: Abundant expression of the interleukin (IL)23 subunit p19, but low levels of bioactive IL23 in the rheumatoid synovium: differential expression and Toll-like receptor-(TLR) dependent regulation of the IL23 subunits, p19 and p40, in rheumatoid arthritis. Ann Rheum Dis 2009, 68:143–150.CrossRefPubMed
17.
Zurück zum Zitat Yeo L, Toellner KM, Salmon M, Filer A, Buckley CD, Raza K, Scheel-Toellner D: Cytokine mRNA profiling identifies B cells as a major source of RANKL in rheumatoid arthritis. Ann Rheum Dis 2011, 70:2022–2028.CrossRefPubMedPubMedCentral Yeo L, Toellner KM, Salmon M, Filer A, Buckley CD, Raza K, Scheel-Toellner D: Cytokine mRNA profiling identifies B cells as a major source of RANKL in rheumatoid arthritis. Ann Rheum Dis 2011, 70:2022–2028.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Salmaggi A, Sandberg-Wollheim M: Monocyte phenotype in blood and cerebrospinal fluid: compartment-specific pattern is unrelated to neurological disease. J Neurol Sci 1993, 120:201–207.CrossRefPubMed Salmaggi A, Sandberg-Wollheim M: Monocyte phenotype in blood and cerebrospinal fluid: compartment-specific pattern is unrelated to neurological disease. J Neurol Sci 1993, 120:201–207.CrossRefPubMed
19.
Zurück zum Zitat Ifergan I, Kebir H, Bernard M, Wosik K, Dodelet-Devillers A, Cayrol R, Arbour N, Prat A: The blood–brain barrier induces differentiation of migrating monocytes into Th17-polarizing dendritic cells. Brain 2008, 131:785–799.CrossRefPubMed Ifergan I, Kebir H, Bernard M, Wosik K, Dodelet-Devillers A, Cayrol R, Arbour N, Prat A: The blood–brain barrier induces differentiation of migrating monocytes into Th17-polarizing dendritic cells. Brain 2008, 131:785–799.CrossRefPubMed
20.
Zurück zum Zitat Cannella B, Raine CS: The adhesion molecule and cytokine profile of multiple sclerosis lesions. Ann Neurol 1995, 37:424–435.CrossRefPubMed Cannella B, Raine CS: The adhesion molecule and cytokine profile of multiple sclerosis lesions. Ann Neurol 1995, 37:424–435.CrossRefPubMed
21.
Zurück zum Zitat Hesse D, Krakauer M, Lund H, Sondergaard HB, Limborg SJ, Sorensen PS, Sellebjerg F: Disease protection and interleukin-10 induction by endogenous interferon-beta in multiple sclerosis? Eur J Neurol 2011, 18:266–272.CrossRefPubMed Hesse D, Krakauer M, Lund H, Sondergaard HB, Limborg SJ, Sorensen PS, Sellebjerg F: Disease protection and interleukin-10 induction by endogenous interferon-beta in multiple sclerosis? Eur J Neurol 2011, 18:266–272.CrossRefPubMed
22.
Zurück zum Zitat Cucci A, Barbero P, Clerico M, Ferrero B, Versino E, Contessa G, Demercanti S, Viglietta E, Di Liberto A, Vai AG, Durelli L: Pro-inflammatory cytokine and chemokine mRNA blood level in multiple sclerosis is related to treatment response and interferon-beta dose. J Neuroimmunol 2010, 226:150–157.CrossRefPubMed Cucci A, Barbero P, Clerico M, Ferrero B, Versino E, Contessa G, Demercanti S, Viglietta E, Di Liberto A, Vai AG, Durelli L: Pro-inflammatory cytokine and chemokine mRNA blood level in multiple sclerosis is related to treatment response and interferon-beta dose. J Neuroimmunol 2010, 226:150–157.CrossRefPubMed
23.
Zurück zum Zitat Calabresi PA, Tranquill LR, McFarland HF, Cowan EP: Cytokine gene expression in cells derived from CSF of multiple sclerosis patients. J Neuroimmunol 1998, 89:198–205.CrossRefPubMed Calabresi PA, Tranquill LR, McFarland HF, Cowan EP: Cytokine gene expression in cells derived from CSF of multiple sclerosis patients. J Neuroimmunol 1998, 89:198–205.CrossRefPubMed
24.
Zurück zum Zitat Hulshof S, Montagne L, De Groot CJ, Van Der Valk P: Cellular localization and expression patterns of interleukin-10, interleukin-4, and their receptors in multiple sclerosis lesions. Glia 2002, 38:24–35.CrossRefPubMed Hulshof S, Montagne L, De Groot CJ, Van Der Valk P: Cellular localization and expression patterns of interleukin-10, interleukin-4, and their receptors in multiple sclerosis lesions. Glia 2002, 38:24–35.CrossRefPubMed
25.
Zurück zum Zitat Navikas V, Link J, Palasik W, Soderstrom M, Fredrikson S, Olsson T, Link H: Increased mRNA expression of IL-10 in mononuclear cells in multiple sclerosis and optic neuritis. Scand J Immunol 1995, 41:171–178.CrossRefPubMed Navikas V, Link J, Palasik W, Soderstrom M, Fredrikson S, Olsson T, Link H: Increased mRNA expression of IL-10 in mononuclear cells in multiple sclerosis and optic neuritis. Scand J Immunol 1995, 41:171–178.CrossRefPubMed
26.
Zurück zum Zitat Ozenci V, Kouwenhoven M, Huang YM, Kivisakk P, Link H: Multiple sclerosis is associated with an imbalance between tumour necrosis factor-alpha (TNF-alpha)- and IL-10-secreting blood cells that is corrected by interferon-beta (IFN-beta) treatment. Clin Exp Immunol 2000, 120:147–153.CrossRefPubMedPubMedCentral Ozenci V, Kouwenhoven M, Huang YM, Kivisakk P, Link H: Multiple sclerosis is associated with an imbalance between tumour necrosis factor-alpha (TNF-alpha)- and IL-10-secreting blood cells that is corrected by interferon-beta (IFN-beta) treatment. Clin Exp Immunol 2000, 120:147–153.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Ozenci V, Kouwenhoven M, Huang YM, Xiao B, Kivisakk P, Fredrikson S, Link H: Multiple sclerosis: levels of interleukin-10-secreting blood mononuclear cells are low in untreated patients but augmented during interferon-beta-1b treatment. Scand J Immunol 1999, 49:554–561.CrossRefPubMed Ozenci V, Kouwenhoven M, Huang YM, Xiao B, Kivisakk P, Fredrikson S, Link H: Multiple sclerosis: levels of interleukin-10-secreting blood mononuclear cells are low in untreated patients but augmented during interferon-beta-1b treatment. Scand J Immunol 1999, 49:554–561.CrossRefPubMed
28.
Zurück zum Zitat Carrieri PB, Provitera V, De Rosa T, Tartaglia G, Gorga F, Perrella O: Profile of cerebrospinal fluid and serum cytokines in patients with relapsing-remitting multiple sclerosis: a correlation with clinical activity. Immunopharmacol Immunotoxicol 1998, 20:373–382.CrossRefPubMed Carrieri PB, Provitera V, De Rosa T, Tartaglia G, Gorga F, Perrella O: Profile of cerebrospinal fluid and serum cytokines in patients with relapsing-remitting multiple sclerosis: a correlation with clinical activity. Immunopharmacol Immunotoxicol 1998, 20:373–382.CrossRefPubMed
29.
Zurück zum Zitat Khademi M, Illes Z, Gielen AW, Marta M, Takazawa N, Baecher-Allan C, Brundin L, Hannerz J, Martin C, Harris RA, Hafler DA, Kuchroo VK, Olsson T, Piehl F, Wallström E: T Cell Ig- and mucin-domain-containing molecule-3 (TIM-3) and TIM-1 molecules are differentially expressed on human Th1 and Th2 cells and in cerebrospinal fluid-derived mononuclear cells in multiple sclerosis. J Immunol 2004, 172:7169–7176.CrossRefPubMed Khademi M, Illes Z, Gielen AW, Marta M, Takazawa N, Baecher-Allan C, Brundin L, Hannerz J, Martin C, Harris RA, Hafler DA, Kuchroo VK, Olsson T, Piehl F, Wallström E: T Cell Ig- and mucin-domain-containing molecule-3 (TIM-3) and TIM-1 molecules are differentially expressed on human Th1 and Th2 cells and in cerebrospinal fluid-derived mononuclear cells in multiple sclerosis. J Immunol 2004, 172:7169–7176.CrossRefPubMed
30.
Zurück zum Zitat Iacobaeus E, Amoudruz P, Strom M, Khademi M, Brundin L, Hillert J, Kockum I, Malmström V, Olsson T, Tham E, Piehl F: The expression of VEGF-A is down regulated in peripheral blood mononuclear cells of patients with secondary progressive multiple sclerosis. PLoS One 2011, 6:e19138.CrossRefPubMedPubMedCentral Iacobaeus E, Amoudruz P, Strom M, Khademi M, Brundin L, Hillert J, Kockum I, Malmström V, Olsson T, Tham E, Piehl F: The expression of VEGF-A is down regulated in peripheral blood mononuclear cells of patients with secondary progressive multiple sclerosis. PLoS One 2011, 6:e19138.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Sawcer S, Hellenthal G, Pirinen M, Spencer CC, Patsopoulos NA, Moutsianas L, Dilthey A, Su Z, Freeman C, Hunt SE, Edkins S, Gray E, Booth DR, Potter SC, Goris A, Band G, Oturai AB, Strange A, Saarela J, Bellenguez C, Fontaine B, Gillman M, Hemmer B, Gwilliam R, Zipp F, Jayakumar A, Martin R, Leslie S, Hawkins S, Giannoulatou E, International Multiple Sclerosis Genetics Consortium Wellcome Trust Case Control Consortium 2, et al.: Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature 2011, 476:214–219.CrossRefPubMedPubMedCentral Sawcer S, Hellenthal G, Pirinen M, Spencer CC, Patsopoulos NA, Moutsianas L, Dilthey A, Su Z, Freeman C, Hunt SE, Edkins S, Gray E, Booth DR, Potter SC, Goris A, Band G, Oturai AB, Strange A, Saarela J, Bellenguez C, Fontaine B, Gillman M, Hemmer B, Gwilliam R, Zipp F, Jayakumar A, Martin R, Leslie S, Hawkins S, Giannoulatou E, International Multiple Sclerosis Genetics Consortium Wellcome Trust Case Control Consortium 2, et al.: Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature 2011, 476:214–219.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Kremlev SG, Gaurnier-Hausser AL, Del Valle L, Perez-Liz G, Dimitrov S, Tuszynski G: Angiocidin promotes pro-inflammatory cytokine production and antigen presentation in multiple sclerosis. J Neuroimmunol 2008, 194:132–142.CrossRefPubMed Kremlev SG, Gaurnier-Hausser AL, Del Valle L, Perez-Liz G, Dimitrov S, Tuszynski G: Angiocidin promotes pro-inflammatory cytokine production and antigen presentation in multiple sclerosis. J Neuroimmunol 2008, 194:132–142.CrossRefPubMed
33.
Zurück zum Zitat Baranzini SE, Elfstrom C, Chang SY, Butunoi C, Murray R, Higuchi R, Oksenberg JR: Transcriptional analysis of multiple sclerosis brain lesions reveals a complex pattern of cytokine expression. J Immunol 2000, 165:6576–6582.CrossRefPubMed Baranzini SE, Elfstrom C, Chang SY, Butunoi C, Murray R, Higuchi R, Oksenberg JR: Transcriptional analysis of multiple sclerosis brain lesions reveals a complex pattern of cytokine expression. J Immunol 2000, 165:6576–6582.CrossRefPubMed
34.
Zurück zum Zitat Lundmark F, Duvefelt K, Iacobaeus E, Kockum I, Wallstrom E, Khademi M, Oturai A, Ryder LP, Saarela J, Harbo HF, Celius EG, Salter H, Olsson T, Hillert J: Variation in interleukin 7 receptor alpha chain (IL7R) influences risk of multiple sclerosis. Nat Genet 2007, 39:1108–1113.CrossRefPubMed Lundmark F, Duvefelt K, Iacobaeus E, Kockum I, Wallstrom E, Khademi M, Oturai A, Ryder LP, Saarela J, Harbo HF, Celius EG, Salter H, Olsson T, Hillert J: Variation in interleukin 7 receptor alpha chain (IL7R) influences risk of multiple sclerosis. Nat Genet 2007, 39:1108–1113.CrossRefPubMed
35.
Zurück zum Zitat Kivisakk P, Matusevicius D, He B, Soderstrom M, Fredrikson S, Link H: IL-15 mRNA expression is up-regulated in blood and cerebrospinal fluid mononuclear cells in multiple sclerosis (MS). Clin Exp Immunol 1998, 111:193–197.CrossRefPubMedPubMedCentral Kivisakk P, Matusevicius D, He B, Soderstrom M, Fredrikson S, Link H: IL-15 mRNA expression is up-regulated in blood and cerebrospinal fluid mononuclear cells in multiple sclerosis (MS). Clin Exp Immunol 1998, 111:193–197.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Lee LF, Axtell R, Tu GH, Logronio K, Dilley J, Yu J, Rickert M, Han B, Evering W, Walker MG, Shi J, de Jong BA, Killestein J, Polman CH, Steinman L, Lin JC: IL-7 promotes T(H)1 development and serum IL-7 predicts clinical response to interferon-beta in multiple sclerosis. Sci Transl Med 2011, 3:93ra68.PubMedPubMedCentral Lee LF, Axtell R, Tu GH, Logronio K, Dilley J, Yu J, Rickert M, Han B, Evering W, Walker MG, Shi J, de Jong BA, Killestein J, Polman CH, Steinman L, Lin JC: IL-7 promotes T(H)1 development and serum IL-7 predicts clinical response to interferon-beta in multiple sclerosis. Sci Transl Med 2011, 3:93ra68.PubMedPubMedCentral
37.
Zurück zum Zitat Vaknin-Dembinsky A, Brass SD, Gandhi R, Weiner HL: Membrane bound IL-15 is increased on CD14 monocytes in early stages of MS. J Neuroimmunol 2008, 195:135–139.CrossRefPubMedPubMedCentral Vaknin-Dembinsky A, Brass SD, Gandhi R, Weiner HL: Membrane bound IL-15 is increased on CD14 monocytes in early stages of MS. J Neuroimmunol 2008, 195:135–139.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Gomez-Nicola D, Spagnolo A, Guaza C, Nieto-Sampedro M: Aggravated experimental autoimmune encephalomyelitis in IL-15 knockout mice. Exp Neurol 2010, 222:235–242.CrossRefPubMed Gomez-Nicola D, Spagnolo A, Guaza C, Nieto-Sampedro M: Aggravated experimental autoimmune encephalomyelitis in IL-15 knockout mice. Exp Neurol 2010, 222:235–242.CrossRefPubMed
39.
Zurück zum Zitat Selmaj K, Raine CS, Cannella B, Brosnan CF: Identification of lymphotoxin and tumor necrosis factor in multiple sclerosis lesions. J Clin Invest 1991, 87:949–954.CrossRefPubMedPubMedCentral Selmaj K, Raine CS, Cannella B, Brosnan CF: Identification of lymphotoxin and tumor necrosis factor in multiple sclerosis lesions. J Clin Invest 1991, 87:949–954.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Matusevicius D, Navikas V, Soderstrom M, Xiao BG, Haglund M, Fredrikson S, Link H: Multiple sclerosis: the proinflammatory cytokines lymphotoxin-alpha and tumour necrosis factor-alpha are upregulated in cerebrospinal fluid mononuclear cells. J Neuroimmunol 1996, 66:115–123.CrossRefPubMed Matusevicius D, Navikas V, Soderstrom M, Xiao BG, Haglund M, Fredrikson S, Link H: Multiple sclerosis: the proinflammatory cytokines lymphotoxin-alpha and tumour necrosis factor-alpha are upregulated in cerebrospinal fluid mononuclear cells. J Neuroimmunol 1996, 66:115–123.CrossRefPubMed
41.
Zurück zum Zitat Buckle GJ, Hollsberg P, Hafler DA: Activated CD8+ T cells in secondary progressive MS secrete lymphotoxin. Neurology 2003, 60:702–705.CrossRefPubMed Buckle GJ, Hollsberg P, Hafler DA: Activated CD8+ T cells in secondary progressive MS secrete lymphotoxin. Neurology 2003, 60:702–705.CrossRefPubMed
42.
Zurück zum Zitat Aloisi F, Pujol-Borrell R: Lymphoid neogenesis in chronic inflammatory diseases. Nat Rev Immunol 2006, 6:205–217.CrossRefPubMed Aloisi F, Pujol-Borrell R: Lymphoid neogenesis in chronic inflammatory diseases. Nat Rev Immunol 2006, 6:205–217.CrossRefPubMed
43.
Zurück zum Zitat Bar-Or A, Fawaz L, Fan B, Darlington PJ, Rieger A, Ghorayeb C, Calabresi PA, Waubant E, Hauser SL, Zhang J, Smith CH: Abnormal B-cell cytokine responses a trigger of T-cell-mediated disease in MS? Ann Neurol 2010, 67:452–461.CrossRefPubMed Bar-Or A, Fawaz L, Fan B, Darlington PJ, Rieger A, Ghorayeb C, Calabresi PA, Waubant E, Hauser SL, Zhang J, Smith CH: Abnormal B-cell cytokine responses a trigger of T-cell-mediated disease in MS? Ann Neurol 2010, 67:452–461.CrossRefPubMed
44.
Zurück zum Zitat Sellebjerg F, Jensen J, Ryder LP: Costimulatory CD80 (B7–1) and CD86 (B7–2) on cerebrospinal fluid cells in multiple sclerosis. J Neuroimmunol 1998, 84:179–187.CrossRefPubMed Sellebjerg F, Jensen J, Ryder LP: Costimulatory CD80 (B7–1) and CD86 (B7–2) on cerebrospinal fluid cells in multiple sclerosis. J Neuroimmunol 1998, 84:179–187.CrossRefPubMed
45.
Zurück zum Zitat Sellebjerg F, Jensen J, Jensen CV, Wiik A: Expansion of CD5 - B cells in multiple sclerosis correlates with CD80 (B7–1) expression. Scand J Immunol 2002, 56:101–107.CrossRefPubMed Sellebjerg F, Jensen J, Jensen CV, Wiik A: Expansion of CD5 - B cells in multiple sclerosis correlates with CD80 (B7–1) expression. Scand J Immunol 2002, 56:101–107.CrossRefPubMed
Metadaten
Titel
Cellular sources of dysregulated cytokines in relapsing-remitting multiple sclerosis
Publikationsdatum
01.12.2012
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2012
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/1742-2094-9-215

Weitere Artikel der Ausgabe 1/2012

Journal of Neuroinflammation 1/2012 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

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.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.