8
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Blood transcriptome profiling captures dysregulated pathways and response to treatment in neuroimmunological disease

      discussion
      EBioMedicine
      Elsevier

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Assessing the transcriptome of blood cells yields a comprehensive picture of the expression status of immune-related genes. In the past decade, impressive technological and analytical advances have given rise to novel high-throughput RNA sequencing and high-density microarray platforms. These approaches perform equally well for well-defined transcriptional regions, while some uncertainties remain regarding the quantification of short and low-abundance transcripts [1]. Blood-based omics have accelerated our understanding of the mechanisms underpinning neuroinflammatory conditions and facilitated the evaluation of therapeutic effects. However, in multiple sclerosis (MS), a chronic immune-mediated neurodegenerative disease that is influenced by complex gene-environment interactions [2], the molecular mechanisms are still not clearly understood. The clinical presentation of MS is highly heterogeneous. Most patients (∼90%) have a relapsing-remitting course of MS (RRMS); characterized by reversible episodes of neurological dysfunction. The modern era of disease-modifying drugs (DMDs) in MS began in 1993 with the approval of interferon-beta (IFN-β) for RRMS [3]. Since then, the therapeutic armamentarium to prevent relapses and progression of disability in patients with MS has increased to >10 DMDs, but injectable IFN-β preparations remain an important option worldwide because of the low risk of severe adverse effects and moderate costs. Recombinant IFN-β stimulates the JAK-STAT pathway, which leads to pleiotropic immunomodulatory effects and reduced migration of immune cells to sites of inflammation in the central nervous system [4]. Still, it is not clear to what extent transcriptional changes in the blood may correlate with clinical outcomes to IFN-β therapy. In an article recently published in EBioMedicine, Feng and colleagues could show by leveraging big transcriptome data that long-term IFN-β therapy in MS corrects an abnormal expression signature of immunoregulatory and neuroprotective genes [5]. A key strength of their analysis is the sophisticated study design, which allowed comparison of RRMS patients vs. healthy controls, treated vs. untreated patients, clinically stable vs. clinically active patients, complete vs. partial responders, and long-term vs. short-term gene expression shifts in response to low dose vs. high dose IFN-β regimens. Feng et al. generated a rich dataset and share it with the scientific community: In total, 227 modern Human Transcriptome Arrays (HTA 2.0), each containing >6 million distinct oligonucleotide probes, were employed to quantify the levels of >67,000 transcripts in peripheral blood mononuclear cells. Furthermore, the authors studied a homogeneous single-centre group of patients (followed for ≥5 years), employed well-established pipelines for rigorous quality control and data analysis, and validated selected findings at the RNA and protein level [5]. Approximately 6000 protein-coding genes and 2000 non-coding RNAs were found to be dysregulated in untreated MS patients compared to healthy controls. This dysregulation was no longer apparent after long-term IFN-β treatment. In the short-term (i.e., within one day following IFN-β injection) >1200 genes were altered in expression. In the long-term response, 277 genes distinguished partial responders (with relapse in the follow-up) from complete responders (with no relapse and no disability progression). The differentially expressed genes have been predominantly implicated in immunity, including JAK-STAT and Toll-like receptor signalling, and neuroprotective pathways, including synaptic transmission. This study thus confirms previously identified gene expression signatures of MS [6], [7] and markedly expands the number of putative biomarkers for IFN-β responsiveness [5]. The insights from the study by Feng et al. [5] can be useful in the counselling of MS patients and for comparing the bioactivity profiles of pegylated and biosimilar forms of recombinant IFN-β, which received marketing authorization in some countries more recently [8]. However, open questions remain regarding the individual risk of transition to secondary progressive MS and the generalizability of the RNA expression pattern for distinguishing partial responders, which needs to be evaluated in independent patient cohorts. Moreover, the expression signatures were not related to neuroimaging and routine laboratory parameters and poor responders (i.e., patients who switched to more effective DMDs because of continued disease activity) were excluded from this study. The large microarray dataset can be further exploited to infer dynamics in the composition of different immune cell types [9] and to analyse alternative splicing events [10]. Future studies may employ single-cell or long-read RNA sequencing solutions and integrate multi-omics information. On the other hand, functional studies are needed to investigate the precise roles of the many non-coding transcripts that were not previously known to be modulated by IFN-β. Longitudinal transcriptome analyses of blood cells represent an attractive approach for exploring the complexities of neuroinflammatory diseases and the mechanisms of action of pharmacologic agents. The data by Feng et al. [5] suggest that long-term IFN-β treatment corrects a peripheral immune imbalance in patients with RRMS. Other DMDs presumably affect this aberrant gene expression as well, though in very different ways. A more comprehensive understanding of molecular disease processes may help to improve the monitoring of therapeutic interventions, which will translate into clinical benefits for patients. The advanced ascertainment of biomarker signatures across a variety of diseases also has the potential to explain previously failed treatment strategies (e.g., IFN-β in neuromyelitis optica) and to inform drug repositioning studies. However, the increased volumes of data from the latest generation of microarray and sequencing technologies and other biomedical sources pose new computational challenges regarding data management, mining, and dissemination. Therefore, in order to optimally prevent inflammatory and neuro-axonal damage and reduce the burden of disease, concerted interdisciplinary efforts are paramount to foster personalized treatment decisions. Declaration of Competing Interest MH received speaking fees and travel funds from Bayer HealthCare, Biogen, Novartis, and Teva.

          Related collections

          Most cited references8

          • Record: found
          • Abstract: found
          • Article: not found

          Treatment of multiple sclerosis — success from bench to bedside

          The modern era of multiple sclerosis (MS) treatment began 25 years ago, with the approval of IFNβ and glatiramer acetate for the treatment of relapsing-remitting MS. Ten years later, the first monoclonal antibody, natalizumab, was approved, followed by a third important landmark with the introduction of oral medications, initially fingolimod and then teriflunomide, dimethyl fumarate and cladribine. Concomitantly, new monoclonal antibodies (alemtuzumab and ocrelizumab) have been developed and approved. The modern era of MS therapy reached primary progressive MS in 2018, with the approval of ocrelizumab. We have also learned the importance of starting treatment early and the importance of clinical and MRI monitoring to assess treatment response and safety. Treatment decisions should account for disease phenotype, prognostic factors, comorbidities, the desire for pregnancy and the patient's preferences in terms of acceptable risk. The development of treatment for MS during the past 25 years is a fantastic success of translational medicine.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            RNA sequencing and transcriptome arrays analyses show opposing results for alternative splicing in patient derived samples

            Background RNA sequencing (RNA-seq) and microarrays are two transcriptomics techniques aimed at the quantification of transcribed genes and their isoforms. Here we compare the latest Affymetrix HTA 2.0 microarray with Illumina 2000 RNA-seq for the analysis of patient samples - normal lung epithelium tissue and squamous cell carcinoma lung tumours. Protein coding mRNAs and long non-coding RNAs (lncRNAs) were included in the study. Results Both platforms performed equally well for protein-coding RNAs, however the stochastic variability was higher for the sequencing data than for microarrays. This reduced the number of differentially expressed genes and genes with predictive potential for RNA-seq compared to microarray data. Analysis of this variability revealed a lack of reads for short and low abundant genes; lncRNAs, being shorter and less abundant RNAs, were found especially susceptible to this issue. A major difference between the two platforms was uncovered by analysis of alternatively spliced genes. Investigation of differential exon abundance showed insufficient reads for many exons and exon junctions in RNA-seq while the detection on the array platform was more stable. Nevertheless, we identified 207 genes which undergo alternative splicing and were consistently detected by both techniques. Conclusions Despite the fact that the results of gene expression analysis were highly consistent between Human Transcriptome Arrays and RNA-seq platforms, the analysis of alternative splicing produced discordant results. We concluded that modern microarrays can still outperform sequencing for standard analysis of gene expression in terms of reproducibility and cost. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3819-y) contains supplementary material, which is available to authorized users.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Systematic review of genome-wide expression studies in multiple sclerosis

              Introduction The aetiology of multiple sclerosis (MS) is complex and involves both genetic susceptibility and environmental factors. However, apart from the widely replicated association with human leukocyte antigen (HLA)-DRB1*1501, genetic risk factors have remained unknown until genome-wide association studies (GWAS), which have recently led to identification of common MS risk variants in over a dozen loci.1–6 Although further fine-mapping and functional studies are required in order to verify the causal variants and genes, a strong presence of immunological genes in these loci is evident. Expression and functional studies in immune cells can therefore elucidate the molecular mechanisms behind MS. Indeed, a number of studies have been conducted where genome-wide expression profiles in peripheral immune cells were compared between MS patients and unaffected controls. Together, these studies have reported a large number of genes with differential expression in MS. However, given that most of these studies have been conducted in small samples without replication, it is likely that many of the findings are false positives. Approaches are therefore needed to increase the probability of detecting the true signals from the vast number of reported genes. In order to extract the genes which are more likely to be true positives, we systematically reviewed results from seven microarray studies in MS, including our previously unpublished study. First, we identified genes which had been found to be differentially expressed in MS to the same direction in at least two studies. In order to further examine the potential role of these most frequently reported genes, they were analysed using pathway tools. Finally, we searched these genes for evidence of association by making comparisons with top results from recent MS GWAS. Materials and methods Samples in the Finnish microarray experiment Twelve female patients fulfilling Poser's criteria for clinically definite MS were recruited through the Seinäjoki Central Hospital. Fifteen healthy unrelated female controls were obtained from the Finnish Twin Study on Ageing (FITSA). The mean age was 54.2 in patients and 71.6 in controls. One patient was receiving cortisone treatment, two patients received β interferon, and one patient was being treated with both β interferon and cortisone at the time of sample collection. All subjects had provided their informed consent. Peripheral blood mononuclear cells (PBMCs) were isolated from whole blood using BD Vacutainer CPT Cell Preparation Tubes (Becton, Dickinson and Company, Franklin Lakes, New Jersy), and cells were disrupted and RNA extracted with TRIzol Reagent (Invitrogen, Carlsbad, California). RNA was then purified using Rneasy Mini Kit (Qiagen, Hilden, Germany), and the sample quality was examined using BioAnalyzer (Agilent, Santa Clara, California). The study was approved by the Committee on Ethics of the Central Hospital of Central Finland and by the Helsinki University Hospital Ethical Committee of Ophthalmology, Otorhinolaryngology, Neurology and Neurosurgery (permit 192/E9/02) for FITSA and patient samples, respectively. Sample processing and microarrays Eleven patient samples were prepared for hybridisation on the Affymetrix GeneChip Human Genome U133 Plus 2.0 Array (Affymetrix, Santa Clara, California) according to the manufacturer's recommendations in our laboratory. In addition, one patient sample and technical replicates from two of the 11 patient samples were prepared according to the manufacturer's recommendations at the Helsinki Biomedicum Biochip Centre (BBC), where 15 control samples had been previously prepared. In brief, 1–2 μg of total RNA was converted to biotin-labelled cRNA using the Affymetrix HT One-Cycle cDNA Synthesis Kit and the HT IVT Labelling Kit. Fifteen micrograms of cRNA was then fragmented and hybridised for 16 h at 45°C, washed in Affymetrix Fluidics Station 450 and scanned with Affymetrix GeneChip Scanner 3000. Hybridisation, washing, staining and scanning were conducted using the same instruments for all samples. All arrays had a present call percentage >40 (42–47) and average background signal 1.4-fold difference in both replicate pairs (N=1668). Finally, we excluded probe sets where the signal in all three MS arrays prepared at the BBC ranked among the four lowest or four highest among the MS arrays (N=2469), after which 15 273 probe sets remained for analyses. After filtering, we discarded the two replicate arrays prepared in our laboratory and used only the replicates prepared at the BBC for final analyses, which thereby included 12 MS arrays and 15 control arrays. In order to identify genes with differential expression in MS, we first applied the fold-change filter in GeneSpring 7.3 using 1.5 as threshold. For probe sets showing a ±≥1.5-fold difference in mean expression between MS patients and controls, we further determined non-parametric Mann–Whitney sum rank test p values with Benjamini–Hochberg correction for multiple testing. Probe sets with corrected a p value of ≤0.05 were considered to be differentially expressed. In order to annotate these probe sets, we compared the gene symbol obtained from GeneSpring 7.3 in October 2008 with the NetAffx annotation in December 2010 for each probe set (http://www.affymetrix.com/analysis/index.affx). If these were different, we first checked whether these were alternative identifiers for the same gene. If not, we obtained the probe set nucleotide sequences from NetAffx and performed a Blat search in UCSC Genome Browser (hg19) to identify the correct target gene (http://genome.ucsc.edu/cgi-bin/hgGateway). Probe sets which recognise only intronic sequences or map to several loci were excluded from the list of differentially expressed probe sets. The Gene Expression Omnibus (GEO) accession number for the microarray dataset is GSE21942 (http://www.ncbi.nlm.nih.gov/geo/). Pathway analysis Pathway analyses were conducted using the Core Analysis option in the Ingenuity Pathway Analysis software (Ingenuity Systems, Redwood City, California). This option identifies canonical pathways associated with a given list of genes by calculating the Fisher exact test p value for the probability that association between this set of genes and a canonical pathway is explained by chance alone. In order to account for the fact that our input lists of genes were enriched for immunological genes and would therefore show association with immune-related pathways if compared with all genes, we restricted the analyses to genes expressed in immune cells by applying the Tissues & Cell lines filter in the analysis settings. Systematic review of microarray studies in MS We searched PubMed with keywords ‘multiple sclerosis microarray’ in November 2010 and obtained 156 records. These were complemented with two additional recent studies and studies identified through a review article.7–9 Based on title, abstract and, if required, full text, we first identified all studies which had been conducted in peripheral immunological cells using a microarray platform and compared expression profiles between MS patients and unaffected controls. This led to the exclusion of 144 studies, which were not MS-related expression studies or investigated effects of MS treatments, were conducted in animal models for MS, or were performed in MS brain biopsies rather than immune cells. We then reviewed the remaining studies in further detail and excluded six studies with fewer than 10 MS patients and/or controls, a study which did not identify any differentially expressed genes, a study where only genes involved in T-cell mediated cytotoxicity were included in the analyses and a study where the list of identified genes was not available in the publication or upon contact with the corresponding author. The stages of the selection process are depicted as a flow chart in figure 1. Of the remaining eight studies which are listed in table 1, only six were independent, because the two studies by Satoh et al 15 16 and the studies by Achiron et al 10 and Mandel et al 13 had been conducted in the same set of patients and were from hereon considered to be single studies. After including our own unpublished experiment, we therefore had seven independent studies for the analysis. For each study, we listed all genes which had been reported to be differentially expressed in MS patients in comparison with healthy controls, and recorded whether their expression in MS was increased or decreased. The studies by Achiron et al 10 and Mandel et al 13 listed only a selected subset of the identified differentially expressed genes.10 13 The corresponding author was contacted in order to obtain the full lists of differentially expressed genes, but we received no reply, and we therefore only included the reported genes. All gene symbols were mapped to the Human Gene Nomenclature gene symbols, and genes that were not unambiguously linked to a single gene symbol using the information available were excluded. Figure 1 Flow chart showing stages in selecting studies for systematic review. MS, multiple sclerosis. Table 1 Description of previous microarray expression studies included in the systematic review Cells Platform No of cases (females/males) No of controls (females/males) Mean age (cases/controls) No of reported differentially expressed genes with a Human Gene Nomenclature symbol Original publication PBMC U95Av2 array (Affymetrix, California). Represents approximately 10 000 genes. 26 (20/6) 18 (16/2) 41/39.6 34 Achiron et al 10 Whole blood 10.5 K Peter MacCallum array (The Peter MacCallum Cancer Institute, Australia). Represents 9381 unique cDNAs. 20 (11/9) Pooled sample (20/0); five individual samples (0/5) Not provided (range 30–66 in MS, 20–55 in controls) 2217 Arthur et al 11 PBMC cDNA arrays with 6500 or 7500 clones with partial overlap 24 (15/9) 19 (5/14) 42/37.5 (age for four controls unknown) 104 Bomprezzi et al 12 Whole blood HT-12 array (Illumina, California). Represents 48 804 transcripts. 99 (66/33) 45 (29/16) 54/48.5 779 Gandhi et al 9 PBMC U95Av2 array (Affymetrix, California). Represents approximately 10 000 genes. 13 (9/4) 5 (4/1) systematic lupus erythematosus (SLE), 10 healthy controls (16/2) 40.7/42.8 (SLE)/39.6 78 Mandel et al 13 PBMC (monocytes depleted) GeneFilters GF211 DNA array (Research Genetics, Alabama). Represents 5184 genes. 15 (11/4) 15 (gender matched to cases, but numbers not available) 42.1/41.7 28 Ramanathan et al 14 T cells, non-T cells A custom array with 1258 cDNAs (Hitachi Life Science, Saitama, Japan) 72 (55/18) 22 (16/6) 36.1/38.6 185 Satoh et al 15 T cells A custom array with 1258 cDNAs (Hitachi Life Science, Saitama, Japan) 72 (55/18) 22 (16/6) 36.1/38.6 281 Satoh et al 16 PBMC, peripheral blood mononuclear cell. Results The previously unpublished microarray screen identifies 692 probe sets with differential expression in MS We first analysed the data from our previously unpublished Finnish microarray screen and identified 692 probe sets, which showed a ≥1.5-fold difference in mean expression between 12 MS patients and 15 controls together with a non-parametric Mann–Whitney sum rank test p value of ≤0.05 after Benjamini–Hochberg correction (supplementary table 1). Three hundred and one probe sets showed increased expression, and 391 decreased expression in MS. Pathway analysis revealed that the differentially expressed genes were strongly associated with PI3K signalling in B lymphocytes (p=1.3E–06), and B cell development (p=2.6E–06) pathways. In addition, altered T cell and B cell signalling in rheumatoid arthritis, role of PKR in interferon induction and antiviral response, and production of nitric oxide and reactive oxygen species in macrophages pathways showed evidence of association with the differentially expressed genes (p 0.8) correlated significantly with the expression of the corresponding DEG (p≤0.001). We also investigated the expression of these genes in risk allele carriers versus non-carriers in lymphoblastoid cell lines of 60 Centre d'Étude du Polymorphisme Humain (CEPH)-derived HapMap samples (CEU) (GEO dataset GSE5859)20 and obtained genotypes for the most strongly associated SNP, or SNPs if identified in several GWASs, from HapMap Release 24 (http://www.hapmap.org).21 In CXCR4, we tested rs882300, which was the most strongly associated CXCR4 SNP in a meta-analysis, instead of the two SNPs mapping within 100 kb of the gene. We did not identify any significant differences in expression between risk allele carriers and non-carriers after correcting for multiple testing. However, we found suggestive evidence for a higher expression of CXCR4 in carriers of the associated G allele at rs882300 (uncorrected one-sided p value=0.04, fold change=1.36) (figure 3), which is in concordance with the higher expression of CXCR4 observed in MS in our Finnish microarray study and three other studies, including two which were excluded from the systematic review owing to small sample sizes.11 22 23 This effect was also seen with the same probe set (217028_at) in our microarray data in a combined sample of 11 MS patients and 14 controls for which a genotype was available (fold change 1.19, one-sided Mann–Whitney test p value=0.06). However, no significant difference in expression levels was observed in two other probe sets measuring for the expression of CXCR4 in our microarray data. Both of these probe sets recognize exonic sequences, while 217028_at identifies 3′UTR in CXCR4. Figure 3 Box plot of CXCR4 expression and rs882300 genotype in 60 Centre d'Étude du Polymorphisme Humain (CEPH) lymphoblastoid cell samples. Discussion Despite extensive research and recent successes in identifying genetic risk variants predisposing to MS, the underlying molecular mechanisms still remain poorly understood. Given the suggested role of autoimmunity and predominance of immunological genes in loci associated with MS, genome-wide expression profiling in immune cells is a valid approach for further elucidating genes and pathways involved in the disease pathogenesis. Although several microarray experiments in MS have been conducted, and a large number of differentially expressed genes have been reported, in most cases the samples have been small, and replication has been lacking, making the findings difficult to interpret. While no obvious consistencies have emerged from these studies, there have not been any systematic attempts to evaluate the overlap between them. We therefore conducted a systematic review of seven microarray studies including our previously unpublished study and confirmed that the general overlap between the studies was indeed poor: only 229 of all 3574 (6%) genes reported to be differentially expressed in MS had been identified in at least two studies, and 94% were therefore unique to a single study. Only 12 of the 229 DEGs were identified in at least three studies. These include NEAT1, which encodes for a non-coding RNA and suppresses the expression of CIITA, an activator of genes within the major histocompatibility complex (MHC) class II locus.24 Interestingly, the most frequently reported gene, HSPA1A, which showed decreased expression in MS in four studies, is also functionally connected to the MHC: it encodes for a heat shock protein, which is likely to be involved in MHC class I and II mediated antigen presentation.25 The gene itself is located in the MHC class III region next to a highly homologous HSPA1B gene, and the measured expression levels may reflect the expression of both genes. However, as is the case for expression studies in general, the studies are not directly comparable owing to differences in samples, sample sizes and platforms, as well as in criteria used for data quality control and for declaring differential expression. Furthermore, the majority of all reported genes came from the only two studies conducted in whole blood samples, and it is therefore not necessarily surprising that most of these genes were not identified in studies conducted in PBMCs or lymphocyte populations. Two studies also reported only a subset of the identified genes, and some studies were conducted using microarrays covering only a fraction of currently known human genes. However, perhaps the most likely explanation for poor overlap across studies is a high rate of false positives and low power to detect true differences in small samples. Recent GWASs conducted in large samples have proven that most of the early genetic associations reported in candidate gene studies of at most a few hundred individuals seem to have been false positives. Small samples may be even more problematic in expression studies, which are susceptible to noise introduced by technical and biological factors. Large studies are required, especially if the aim is to identify expression changes which are due to genetic disease risk variants because the effects of common genetic variants on gene expression are in most cases relatively modest, even in rather homogeneous cell populations,26 and in small samples the difference in risk allele frequency between cases and controls is not expected to be significant in the first place. However, after excluding 13 genes in the MHC region, the 229 in silico replicated DEGs were enriched for variants showing a modest association in the IMSGC GWAS,1 suggesting that at least some of these genes are likely to play a causative role in MS rather than showing differential expression as a result of activation of immunological pathways secondary to MS. In addition, 15 of these genes have shown suggestive evidence for association with MS (p<0.0001) including CDK4, IL7R and TNFRSF1A, which are located in regions of genome-wide significance.1 3 5 17 These 15 also include CD40, which is associated with rheumatoid arthritis,27 and TNFAIP3, which is associated with coeliac disease, SLE, psoriasis and rheumatoid arthritis.28–31 TNFAIP3 was also one of the genes identified as differentially expressed in three studies, including our own experiment.11 15 16 It encodes for a zinc finger protein which inhibits nuclear factor (NF)-κβ activity and tumour necrosis factor (TNF)-mediated programmed cell death, and may therefore play an important role in regulating various immunological pathways.32 However, apart from CXCR4, the expression of the 15 DEGs did not correlate with the proposed risk variants in lymphoblastoid cell lines, although potential eQTL effects should be further investigated in other immune cells populations. In CXCR4, the associated SNP correlated modestly with expression when measured by a probe set representing the 3′UTR, which may indicate differential usage of alternative polyadenylation signals. Interestingly, CXCR4 promotes transendothelial migration of T cells in vitro together with its ligand, CXCL12,33 while CXCR4 and CXCR3 antagonists reduce the accumulation of CD4+ T cells in the CNS and inhibit EAE pathogenesis.34 We acknowledge that results from our previously unpublished experiment may have been affected by technical factors as well as by the age difference between cases and controls. The study also included four patients who had received treatment. We therefore reviewed the list of DEGs after excluding our study and found that 135 of the 229 DEGs were identified in at least two independent studies. Pathway analysis on both the 229 and 135 in silico replicated DEGs showed that they were highly associated with several immunological pathways. Interestingly, the identified interleukin signalling pathways (IL-4, IL-6 and IL-17) are primarily related to Th2 and Th17 cells rather than Th1 cells, which are thought to mediate MS.35–37 Further, IL-6 regulates the balance between regulatory T cell and Th17 cell differentiation together with TGF-β.38 Th17 cells have been linked with autoimmunity, and several studies have provided evidence for their role in MS and EAE,39 while regulatory T cells have been demonstrated to display loss of suppressive function in MS.40 Further studies are needed to investigate whether changes in expression of genes in these interleukin signalling pathways are causative or secondary to MS. We also saw a strong association with cancer-related pathways, which may suggest some common molecular mechanisms behind cancer and autoimmunity, such as dysregulation of apoptosis signalling. Finally, the pathway showing most significant association with the in silico replicated DEGs was the glucocorticoid receptor signalling pathway, which is a central regulator of inflammation. Although one could speculate that this may reflect the usage of corticosteroids as a treatment for MS, patients in the included studies had reportedly been untreated shortly prior to sample collection apart from our study where two patients had been treated with cortisone. This pathway was also the most significantly associated when our study, which included treated patients, was excluded. This would suggest that the regulation of endogenous glucocorticoid receptor signalling pathway may be disturbed in MS, which is in concordance with previous evidence of reduced glucocorticoid receptor binding affinity and sensitivity in lymphocytes in MS patients.41 42 Furthermore, mice producing an antisense RNA for the glucocorticoid receptor do not develop EAE.43 Interestingly, several of the genes in confirmed MS risk loci are connected to the glucocorticoid receptor signalling pathway, including STAT3, which was recently identified through a GWAS by our group4 and acts as a co-activator of glucocorticoid receptor signalling.44 In conclusion, we have performed the first systematic review of microarray studies in MS. In general, there was little overlap between the seven studies investigated, most likely owing to the small sizes of these studies. However, 229 genes were reported to be differentially expressed in MS in at least two studies. After excluding our unpublished experiment, which may have been affected by confounding factors and inclusion of treated subjects, 135 genes were identified in at least two studies. Of the 229 genes, 12 were reported in at least three studies, including TNFAIP3, which is associated with several other autoimmune diseases, and NEAT1 and HSPA1A, which are both functionally connected to MHC, the major MS susceptibility locus. Pathway analyses on the 229 and 135 DEGs provided support for the involvement of glucocorticoid receptor signalling and Th2, Th17 and regulatory T-cell-related interleukin signalling pathways in MS. Together with accumulating data from genetic association studies, our findings can be helpful in selecting genes and pathways for further functional studies in MS. Supplementary Material Supporting Statement Author's manuscript Reviewer comments
                Bookmark

                Author and article information

                Contributors
                Journal
                EBioMedicine
                EBioMedicine
                EBioMedicine
                Elsevier
                2352-3964
                24 October 2019
                November 2019
                24 October 2019
                : 49
                : 2-3
                Affiliations
                [0001]Rostock University Medical Center, Department of Neurology, Division of Neuroimmunology, Gehlsheimer Str. 20, 18147 Rostock, Germany
                Article
                S2352-3964(19)30705-4
                10.1016/j.ebiom.2019.10.035
                6945196
                31668881
                b648b387-fdeb-4059-8233-c173cf56d720
                © 2019 The Author. Published by Elsevier B.V.

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 18 October 2019
                : 18 October 2019
                Categories
                Commentary

                Comments

                Comment on this article