Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Feb 1.
Published in final edited form as: Mult Scler Relat Disord. 2018 Dec 5;28:34–43. doi: 10.1016/j.msard.2018.11.032

Cerebrospinal Fluid Biomarkers Link Toxic Astrogliosis and Microglial Activation to Multiple Sclerosis Severity

Ruturaj Masvekar 1, Tianxia Wu 2, Peter Kosa 1, Christopher Barbour 1,3, Valentina Fossati 4, Bibiana Bielekova 1,*
PMCID: PMC6411304  NIHMSID: NIHMS1516620  PMID: 30553167

Abstract

Background:

Once multiple sclerosis (MS) reaches the progressive stage, immunomodulatory treatments have limited efficacy. This suggests that processes other than activation of innate immunity may at least partially underlie disability progression during late stages of MS. Pathology identified these alternative processes as aberrant activation of astrocytes and microglia, and subsequent degeneration of oligodendrocytes and neurons. However, we mostly lack biomarkers that could measure central nervous system (CNS) cell-specific intrathecal processes in living subjects. This prevents differentiating pathogenic processes from an epiphenomenon. Therefore, we sought to develop biomarkers of CNS cell-specific processes and link them to disability progression in MS.

Methods:

In a blinded manner, we measured over 1000 proteins in the cerebrospinal fluid (CSF) of 431 patients with neuroimmunological diseases and healthy volunteers using modified DNA-aptamers (SOMAscan®). We defined CNS cell type-enriched clusters using variable cluster analysis, combined with in vitro modeling. Differences between diagnostic categories were identified in the training cohort (n = 217) and their correlation to disability measures were assessed; results were validated in an independent validation cohort (n = 214).

Results:

Astrocyte cluster 8 (MMP7, SERPINA3, GZMA and CLIC1) and microglial cluster 2 (DSG2 and TNFRSF25) were reproducibly elevated in MS and had a significant and reproducible correlation with MS severity suggesting their pathogenic role. In vitro studies demonstrated that proteins of astrocyte cluster 8 are noticeably released upon stimulation with proinflammatory stimuli and overlap with the phenotype of recently described neuro-toxic (A1) astrocytes.

Conclusion:

Microglial activation and toxic astrogliosis are associated with MS disease process and may partake in CNS tissue destruction. This hypothesis should be tested in new clinical trials.

Keywords: multiple sclerosis, CSF biomarkers, microglial activation, toxic astrogliosis, CNS tissue destruction, MS severity

Graphical Abstract

graphic file with name nihms-1516620-f0001.jpg

1). INTRODUCTION

Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system (CNS), in which immune cells induce demyelination and axonal damage. MS pathogenesis has been partially understood (Mohajeri et al., 2015). The disease course and prognosis are largely unpredictable (Roxburgh et al., 2005; Weideman et al., 2017a), but once MS reaches progressive stage, current disease-modifying treatments (DMTs) have limited efficacy (Hartung et al., 2002; Kappos et al., 2001; Montalban et al., 2016; Weideman et al., 2017b).

These data suggest that cells of the adaptive immunity play a pivotal role in initiating the MS disease process, with T- and B-lymphocytes, and plasma cells found abundant in the early MS lesions (Frischer et al., 2009; Kirk et al., 2003; Lassmann and van Horssen, 2011). But in later stages of MS, formation of new lesions decreases and may completely stop. Furthermore, focal lesions in progressive MS contain generally low numbers of T-cells and activated microglia/macrophages (Frischer et al., 2009; Kroksveen et al., 2015; Revesz et al., 1994). Consequently, inflammation was thought to no longer drive disability accumulation in progressive MS. In fact, primary-progressive MS was considered a non-inflammatory disease (Stys et al., 2012). However, MS patients from all three phenotypical subtypes (i.e., relapsing-remitting [RR-MS], secondary- [SP-MS], and primary-progressive MS [PP-MS]) were found to have identical concentrations of cerebrospinal fluid (CSF) biomarkers specific for B cells (sCD21) or T cells (sCD27) (Komori et al., 2015). The conclusion that patients with progressive MS have identical levels of inflammation that is redistributed outside of MS lesions visible on MRI, e.g. to meninges (Choi et al., 2012; Magliozzi et al., 2007; Reynolds et al., 2011) or normal-appearing CNS tissue (Kutzelnigg et al., 2005), is strongly supported by pathology data.

Nevertheless, the mere presence of the immune cells in CNS does not prove their pathogenicity in progressive MS, where pathologist described alternative processes such as hypoxia (Lassmann, 2016), mitochondrial dysfunction (Mahad et al., 2008) and endoplasmic reticulum stress (McMahon et al., 2012), affecting both neurons and oligodendrocytes. Most recently, a specific type of astrocyte activation, called “neurotoxic reactive astrocytes (or “A1” astrocytes)” was observed in several neurodegenerative diseases, including MS (Liddelow et al., 2017a). If these alternative processes contribute to disability progression, then they must be therapeutically-targeted if we hope to attain greater than 25% decrease of disability progression achievable by current treatments (Montalban et al., 2016) in progressive MS.

However, the inability to measure aforementioned intrathecal processes in living subjects impedes development of treatments that may inhibit them. It is likely that the presence or dominance of such potentially pathogenic processes varies among individual patients, depending on their genetic background and environmental influences. Thus, patients enrolled in clinical trials include subjects with and without targeted process, diminishing the probability of proving efficacy of process-targeted treatment in early clinical testing. We draw an analogy to cardiovascular diseases, where several processes, such as hypertension, hypercoagulable states, hyperlipidemia and diabetes have proven pathogenicity on a group level, but each is present only in a proportion of affected patients. Without biomarker-based pre-selection of suitable patients for clinical trials, the efficacy of process-specific treatments (e.g. anti-hypertensive or diabetic drugs) in cardiovascular diseases would be substantially lower. Furthermore, we could not assemble drugs into logical, highly-efficacious patient-specific combinations in daily clinical practice.

Thus, there is a need to identify and validate biomarkers reflective of CNS cell-specific processes. In a previous publication, we focused on immune cells and defined CSF biomarkers that can reliably quantify intrathecal inflammation while also providing information about its cellular composition (Komori et al., 2015). Here, we sought to take an analogous approach to identify biomarkers that are predominantly secreted by specific CNS cells, assemble related biomarkers into clusters to decrease dimensionality and to investigate whether concentrations of such CNS cell-enriched clusters differ in MS patients during disease evolution. Finally, we asked whether these clusters correlate with clinical measures of disability and MS severity.

2). MATERIALS and METHODS

2.1). Research subjects

All subjects (n = 431) were prospectively recruited under National Institutes of Health (NIH) Central CNS Institutional Review Board-approved protocol (Comprehensive Multimodal Analysis of Neuroimmunological Diseases of the Central Nervous System; ClinicalTrials.gov Identifier: NCT00794352) and provided informed consent. All subjects underwent detailed neurological examination that included clinician-derived disability scales: Expanded Disability Status Scale (EDSS(Kurtzke, 1983)) and Scripps Neurological Rating Scale (SNRS(Sipe et al., 1984)). Functional disability scales: 25-foot walk (25FW) and nine-hole peg test (9HPT) were collected by non-clinical investigators at the same visits. These four disability measures were then assembled to sensitive Combinatorial Weight-Adjusted Disability Scale (CombiWISE (Kosa et al., 2016)). Additionally, clinical-grade structural MRI images of the brain collected at 1.5T or 3T MRIs under published protocols (Kosa et al., 2015) were semi-quantitatively graded by trained clinical investigators to derive Composite MRI scale of CNS tissue destruction (COMRIS-CTD(Kosa et al., 2015)). The detailed treatment information (i.e. initiation and termination dates for all MS disease-modifying treatments (DMTs)) were assembled in research database. EDSS-based MS severity scale (Multiple Sclerosis Severity Score (MSSS)) (Roxburgh et al., 2005) and Age Related Multiple Sclerosis Severity (ARMSS) (Manouchehrinia et al., 2017) were calculated using published algorithms. Clinical, MRI and therapeutic history data were used to calculate Multiple Sclerosis Disease Severity Scale (MS-DSS) according to published algorithm (Weideman et al., 2017a) that is available on public website: https://bielekovalab.shinyapps.io/msdss/. This website also includes computation of CombiWISE and COMRIS-CTD scales. MS-DSS has significantly enhanced sensitivity and specificity in comparison to MSSS and ARMSS and can predict future rates of disability progression in an independent cohort.

Before analyses, subjects were randomly divided into two cohorts (Table 1): training (n = 217) and validation cohort (n = 214). The cohorts were split in a way that kept gender ratio, age spread, disease diagnosis and disease disability/severity scales comparable for both cohorts. Patients were first binned into groups based on diagnosis and whether CombiWISE scores were available. These groupings that had 10 or more patients were further split by gender. Within each of these groupings, patients were further binned as per whether they were above or below the median measurements of age and CombiWISE (when present). Approximately 50% of patients in each of these final groupings were randomly selected as the training cohort, with the remaining 50% retained as the validation cohort.

Table 1:

All subjects’ (n = 431) were divided into two cohorts: Training Cohort (n = 217) and Validation Cohort (n = 214). Gender-ratio, age-, disease diagnostics groups-, disease associated disability scores-range were comparable in the two cohorts.

Training Cohort(N=217) Validation Cohart (N=214)

Diagnosis HD NIND CIS RRMS SPMS PPMS HD NIND CIS RRMS SPMS PPMS
N (female/male) 9/10 20/8 8/2 41/25 21/17 24/32 11/12 23/6 7/3 38/23 21/13 28/29

Average 39.3 43.9 39.7 39.6 51.2 53.6 35.8 49.2 41.0 39.6 53.3 54.3
Age (SD) 15.4 12.8 14.4 10.9 11.6 9.5 10.5 11.2 11.8 10.5 8.8 9.0
range 19.4–70.3 18.2–65.8 20.8–60.3 18.0–68.7 22.0–65.4 25.3–65.8 19.7–57.4 21.0–70.6 24.4–57.7 18.3–67.9 36.3–69.6 29.8–74.7

Average 0.4 2.7 1.4 2.0 5.6 5.3 0.4 2.0 2.0 1.5 5.8 5.0
EDSS (SD) 0.5 2.3 1.1 1.5 1.7 1.8 0.5 2.0 0.9 1.0 1.3 1.7
range 0.0–1.5 0.0–6.5 0.0–3.0 0.0–6.5 0.0–8.0 1.5–8.5 0.0–1.0 0.0–6.5 1.0–4.0 0.0–6.0 2.5–7.5 1.5–7.0

Average 5.4 19.9 11.9 15.2 46.7 41.0 4.9 16.1 15.4 12.5 46.5 39.9
CombiWISE (SD) 3.6 16.4 7.0 10.3 13.1 15.4 2.9 14.0 5.7 6.7 13.2 13.8
range 0.4 12.2 1.2–55.6 2.6 22.2 2.2 50.4 17.8 80.6 9.4 84.5 0.7–9.5 2.3–53.2 7.5–24.5 2.4 35.5 20.3–70.8 13.8–66.2

Average 1.0 5.9 2.1 8.2 16.7 13.8 1.5 6.9 4.3 8.5 15.7 14.0
COMRIS (SD) 1.7 6.9 1.8 5.5 5.5 6.3 2.7 4.8 3.2 5.4 5.2 6.1
range 0.0–5.6 0.0–25.5 0.0–5.7 0.0–22.0 6.6–25.7 3.7–31.5 0.0–9.2 0.0–21.2 0.0–9.2 0.0–23.5 8.0–25.6 0.0–26.3

Average 1.2 1.5 0.9 1.3 2.3 1.9 1.2 1.4 1.0 1.2 2.3 2.1
MS-DSS (SD) 0.1 0.9 0.2 0.6 1.2 1.1 0.1 0.8 0.3 0.5 1.0 0.9
range 0.8–1.4 0.5–3.8 0.6–1.3 0.4–3.5 0.3–5.3 0.3–4.8 0.8–1.5 0.5–4.4 0.6–1.6 0.5–2.5 0.7–4.5 0.8–4.4

2.2). CSF Collection and Processing

CSF were collected by lumbar puncture and processed as per standard operating procedure as described (Barbour et al., 2017). Briefly, research CSF aliquots were placed on ice immediately after collection, assigned an alpha-numeric code, and centrifuged at 335g for 10 minutes at 4°C, at Neuroimmunological Diseases Section (NDS) laboratory within 15 minutes of collection. CSF supernatants were aliquoted and stored at −80°C until further use.

2.3). PBMCs Isolation, Stimulation, and Supernatants Collection

Peripheral Blood Mononuclear Cells (PBMCs) were isolated from whole blood using Ficoll separation method (Noble and Cutts, 1967). Briefly, lymphocyte separation media (LSM; Lonza, Walkersville, MD) was underlaid to pre-diluted blood (1:3 dilution with phosphate-buffered saline [PBS]; KD Medical Inc., Columbia, MD) and then centrifuged at 335g for 30 min at room temperature. PBMC were collected, washed twice with ice cold PBS, counted by hemocytometer and cultured in serum-free X-VIVO media (Lonza) at 1×106 cells/ml. PBMCs were stimulated with lipopolysaccharide (LPS; 100 ng/ml; Sigma-Aldrich, St. Louis, MO) and CD3/CD28 microbeads (Invitrogen, Carlsbad, CA) at 1:1 cells to beads ratio. After 24h supernatants from stimulated and unstimulated PBMCs were collected by centrifugation at 335g. Stimulation of PBMCs was validated by analyzing pro-inflammatory cytokines in supernatants using Human Pro-Inflammatory 7-Plex Tissue Culture Kit (Meso Scale Discovery, Rockville, MD; Supplementary Figure 1). Supernatants were aliquoted and stored at −80°C until further use.

2.4). CNS Cell Cultures and Treatments

Primary human CNS cells or cell lines: human neurons (ScienCell, Carlsbad, CA), human astrocytes (ScienCell), human brain endothelial cell line (HCMEC/D3; provided by Pierre-Olivier Couraud (Weksler et al., 2005)) and human microglia cell line (CHME5; provided by Nazira El-Hage, Florida International University, Miami, FL) were plated (105 cells/ml) according to manufacturer’s/provider’s instructions. These cells were treated with respective culture medium (Control) or inflammatory stimuli (supernatant from LPS- and CD3/CD28 beads-stimulated PBMCs, 50% volume/volume) or mitochondrial-stress stimulus (rotenone, 100 nM). Cell-culture supernatants were collected at 0 and 24 hours after respective treatments and stored at −80°C until further use. Oligodendrocytes were differentiated from human pluripotent stem cells as described (Douvaras and Fossati, 2015); differentiated cultures had 70% of O4+oligodendrocytes.

2.5). SOMAscan

Cell-culture and CSF supernatants were analyzed using Slow Off-rate Modified DNA Aptamers assay (SOMAscan; SomaLogic Inc., Boulder, CO), a multiplex proteomic scan utilizing modified DNA-aptamers (SOMAmers). Cell-free CSF supernatants were analyzed using 1.1K SOMAscan platform (determines expression of 1128 different proteins, available between June 2012 and October 2016), and Cell-culture supernatants were analyzed using 1.3K SOMAscan platform (determines expression of 1310 different proteins, available after October 2016). Binding of SOMAmers to specific proteins enables transforming individual protein concentration into DNA concentrations quantified by hybridization (Barbour et al., 2017; Gold et al., 2010; Gold et al., 2012; Kraemer et al., 2011; Rohloff et al., 2014).

2.6). Identification of CNS Cell Type-Enriched Biomarkers and Biomarker Clusters

Supernatants collected from primary human CNS cells and cell lines (see Methods) at 0 and 24 hours after culturing were analyzed using SOMAscan. If under control culture conditions absolute release of any protein by a specific cell-type (i.e., RFU at 24h – RFU at 0h) was 5-fold higher than any other cell types then that protein was considered specific cell type-enriched. However, human embryonic stem cells-derived oligodendrocyte cultures had only 70% of O4+oligodendrocytes, with astrocytes and neurons representing the remaining cells in these cultures. To circumvent problem of mixed cultures of oligodendrocytes, we first identified cell type-enriched biomarkers for all CNS cell types except oligodendrocytes. This analysis identified neuron-enriched, astrocyte-enriched, microglia-enriched and endothelial cells-enriched biomarkers. To identify oligodendrocyte-enriched biomarkers, we repeated the same analysis including oligodendrocytes, but considered biomarker to be oligodendrocyte-enriched only when the biomarker was not previously identified as enriched for some other CNS cell type. We recognize that the drawback of this sequential process is that some biomarkers identified as enriched for non-oligodendrocyte cell types may in fact be also produced by some oligodendrocytes. However, due to low numbers of embryonic stem cells-derived oligodendrocyte we could not subject these cultures to second isolation procedure.

CNS cell type-enriched biomarker clusters were obtained by performing variable cluster analysis (see Statistical Analyses) for each cell type-enriched proteins on patients’ CSF SOMAscan data. We reasoned that proteins that are secreted together by identical CNS cell types under identical physiological or pathological conditions will have strong correlations in the CSF samples collected from diverse group of individuals.

2.7). Statistical Analyses

2.7.1). Data transformation and outlier Winsorization (the two cohorts combined):

For each cell type-enriched biomarker, Box-Cox transformation was applied, and kurtosis was examined. If the transformed variable had an absolute kurtosis greater than 2, histogram and interquartile range (IQR) were used to identify extreme outliers based on the 3*IQR rule. The outlier(s) was replaced by either the minimum or maximum after said outlier(s) were excluded.

2.7.2). Variable cluster analysis (the two cohorts combined):

For each cell type-enriched biomarkers variable cluster analysis was performed on patients’ CSF SOMAscan data (after adjusting or excluding outliers), using the SAS procedure VARCLUS (SAS/STAT® 13.2 User’s Guide; SAS Institute Inc., Cary, NC). The maximum second eigenvalue (MAXEIGEN) of 0.7 was used as the criterion for cluster splitting. For each cluster, procedure SCORE was used to calculate the (first principal) component score, which is the weighted average of the variables that explains as much variance in the cluster as possible.

2.7.3). Correlation analysis (separate test for training and validation cohort):

Semi-partial correlation coefficients (Stevens, 2002; Whittaker, 1990) were used to examine for associations between cluster scores and clinical outcomes. These allow for testing of associations between cluster scores and clinical outcomes, after accounting for the association of a confounding variable (i.e. age) with cluster scores. For each cluster, semi-partial Spearman correlation coefficients were calculated using the ppcor R package (Kim, 2015) to evaluate the association between cluster scores (after accounting for potential associations with age) and the clinical outcomes: CombiWISE, EDSS, COMRIS-CTD, MS-DSS, ARMSS and MSSS. In addition, the individual proteins making up two clinically relevant clusters (Astrocyte cluster 8 and Microglial cluster 2) were examined. Only clusters with a statistically significant (p < 0.01) correlation coefficients for at least one clinical outcome in the training cohort were tested in the validation cohort. The correlation p-values in the validation cohort were adjusted in R (2018) using the Benjamini-Hochberg False Discover Rate method (Benjamini and Hochberg, 1995).

2.7.4). Examining for differences in disease subgroups (separate test for training and validation cohort):

For each cluster, a linear regression model with diagnostic subgroup and age as independent variables was constructed. Analysis of Variance (ANOVA) was performed to examine for differences in component scores among diagnostic subgroups after accounting for differences in age. When statistically significant (p-value < 0.05) cluster differences were found, pairwise multiple comparisons using Tukey’s method were performed. When graphically displayed, cluster scores are adjusted for age by subtracting the respective estimated age effects from these linear regression models.

3). RESULTS

3.1). Identification of CNS Cell Type-Enriched Biomarkers

CNS cell type-enriched biomarkers were identified using cell culture supernatants’ SOMAscan data (see Methods for more details). To identify absolute release of any biomarker by a specific CNS cell type, the absolute difference in RFU values between 24h and 0h supernatants was considered to reflect secretion (or consumption) of the measured protein by studied cell type. We arbitrarily defined measured protein as cell type-enriched, if release of a protein by a specific cell type under control culture conditions was 5-fold higher than any other tested cell type. The biomarkers that fulfilled our criteria of >5-fold enrichment by a specific CNS cell type are represented in Supplementary Table 1: 40 neuron-, 73 astrocyte-, 81 oligodendrocyte-, 18 microglia-, and 38 endothelial cell type-enriched biomarkers.

3.2). Identification of CNS Cell Type-Enriched Biomarker Clusters

CNS cell type-enriched biomarker clusters were obtained by performing variable cluster analysis for each cell type-enriched biomarkers (shown in Supplementary Table 1) in patients’ CSF SOMAscan data. Variable cluster analysis resulted in cell type-enriched biomarker clusters, represented in Table 2: 7 neuron-, 20 astrocyte-, 19 oligodendrocyte-, 7 microglia-, and 11 endothelial cell type-enriched biomarker clusters.

Table 2:

CNS cell type-enriched biomarker clusters were obtained by performing variable cluster analysis for each cell type-enriched proteins on patients’ CSF SOMAscan data. Table represents list of CNS cell type-enriched biomarker clusters.

Neuron Cluster 1 IFNG
CMPKl
MASP1
STK17B
PRSS27 OCIAD1 DKKL1 PPP3R1 KLK14 ING1 DAPK2 SH2D1A
Cluster 2 FGFR1
DLL1
CYCS PROC CNTN1 BCAN CHL1 ITGAV RTN4R NOTCH3 IL27RA
Cluster 3 LTBR ARG1 PTPN1 AURKA KLK6 ATP5B
Cluster 4 LTF ALPL
Cluster 5 CAPN1 WISP1 CAMK1 CD84 INS
Cluster 6 C1QBP
Cluster 7 CDH3 EPB41 HPGD
Astrocyte Cluster 1 COL18A1 CXCL16 MMP3 ANGPT1 MRC2 LRIG3 PLAU BDC CLU MAPK11
Cluster 2 BMP1 TNFRSF4 MMP10 FGF23 ADAM12 ADAMTS1 MMP13
Cluster 3 INHBA THBS1 KDR MMP2 GSN OMD
Cluster 4 DYNLRB1 TPM2 ZAP70
Cluster 5 DCN FRZB PTHLH CLEC11A ESM1 TNC CLEC11A DBNL
Cluster 6 ALB C4
Cluster 7 TNFSF13R IL6
Cluster 8 MMP7 SERPINA3 GZMA CLIC1
Cluster 9 ANGPT2 GDF11 ADAM9 PRSS22 NOTCH1 UNC5C APOE SPOCK1
Cluster 10 NOV CCL11
Cluster 11 SPARC
Cluster 12 CAMK2D FGF1
Cluster 13 NOG
Cluster 14 TGFB2
Cluster 15 F10 SERPINA5 INHBA TNFAIP6
Cluster 16 PRKCD BPI GREM1 IL11
Cluster 17 PGD
Cluster 18 LAYN MMP8 IL17RA CA4
Cluster 19 CCL2
Cluster 20 IGFBP5 RARRES2
Oligodendrocyte Cluster 1 EFNB3
NTRK2
APOE
NRCAM
JAM2
RCB02
L5AMP
ICAM5
LRP8
TNFRSF21
HFE2 RGMA L1CAM NCAM1 SUTRK5
Cluster 2 HGF TNFSF15 CD209 FCGR3B LY86 CXCL10 CHST6 TFF3 GPNMB
Cluster 3 EHMT2 PYY CKB FYN
Cluster 4 FTH1_FTL HBA1_HBB
Cluster 5 IGFBP3 PROS1 MATN2 KLKB1 TF DPT SET SERPINA3_ Complex
Cluster S GDF9 COLEC12 PDGFC MFD1 LRRTM3 FABPS
Cluster 7 CTSB SFRP1 NLGN4X SEMA3E SPOCK2
Cluster 8 CDH1 PLA2G2A GPC3
Cluster 9 ICAM1 APOD CCL7 TYK2
Cluster 10 C4B C1R A2M
Cluster 11 HP
Cluster 12 ERBB3
GPC5
LAMA1_LA
CPE
THBS4 PTPN11 AGT DKK3 CTSH IGF1R SPON1 SPARCL1
Cluster 13 CX3CL1
Cluster 14 CXCL13
Cluster 15 APCS
Cluster 16 F5
Cluster 17 SHBG
Cluster 18 PAFAH1B2 AK1
Cluster 19 IDUA
Microglia Cluster 1 VEGFA WFIKKN2 CHRDL1 GPT IL12RB2 FABP3 GOT1 SEMA6A
Cluster 2 DSG2 TNFRSF25
Cluster 3 SEMA3A VEGFA WNT7A
Cluster 4 CTSD
Cluster 5 IGFBP1
Cluster 6 PGK1 PPID IMPDH1
Cluster 7 MIA
Endothelial Cells Cluster 1 TIE1 TGFBR3 VWF TNFRSF1B CST6 IL6R TPI1 CD274 MICB SCARF1
Cluster 2 KPNA2 TOPI C3 EIF4A3 SSRP1
Cluster 3 SERPINA7 PGF C1S CFB F9 F2
Cluster 4 CCL5 IL2RG XRCC6 CDH6 CDH12 H2AFZ CSNK2A1
Cluster 5 DKK4 DKK1 MAPK14
Cluster 6 CST2
Cluster 7 GNS
Cluster 8 TK3
Cluster 9 NAGK
Cluster 10 MICA
Cluster 11 C3 MB

3.3). Analysis of Cluster Scores Across Disease Diagnosis Subgroups

Cluster scores for each biomarker cluster were calculated using the first principal component. After accounting for associations of age with cluster scores (see Statistical Analyses), cluster scores were compared among disease diagnosis subgroups (healthy donors [HD], clinically isolated syndrome [CIS], non-inflammatory neurological disorder [NIND], RR-MS, SP-MS, and PP-MS patients) in the training cohort (n = 217). Statistically-significant differences were then assessed in the independent-validation cohort (n= 214). Only those statistically-significant results that were reproduced in both cohorts are represented and will be discussed here.

All MS subgroups had statistically significantly elevated astrocyte cluster 8 (MMP7, SERPINA3, GZMA and CLIC1; Figure 1, first row) in comparison to HD and NIND subgroups; while astrocyte cluster 11 (SPARC; Figure 1, second row) showed a decrease in SP-MS subgroup compared to NIND. Microglia cluster 2 (DSG2 and TNFRSF25; Figure 1, third row) behaved similarly to astrocyte cluster 8, that it was increased in all 3 MS subtypes in comparison to HD and NIND. Finally, oligodendrocyte cluster 14 (CXCL13; Figure 1, fourth row) demonstrated elevation in RR-MS compared to HD and NIND, while PP-MS was elevated compared to only NIND subgroup.

Figure 1:

Figure 1:

Cluster scores adjusted based on age were compared among diagnosis subgroups. Only clusters which have reproducible statistically significant differences between disease diagnosis subgroups are shown. The y-axis as selected to have better visual assessment of majority of patients’ cluster scores. Thus, few of individual patients’ points may be missing in these graphs. Data were analyzed using ANOVA, with multiple comparisons using Tukey’s method; *p < 0.05, **p < 0.005, ***p < 0.0005 and ****p < 0.0001.

3.4). Correlation Analysis Between Cluster Scores and Disability and CNS Tissue Destruction

To determine if CNS cell-specific biomarkers increase with MS progression, we correlated individual subject cluster scores with two MS disability measures: broadly used EDSS (Kurtzke, 1983) and more sensitive CombiWISE (Kosa et al., 2016). Additionally, we also correlated cluster scores with the amount of CNS tissue destruction measured by COMRIS-CTD (Kosa et al., 2015) using semi-partial Spearman correlations that is generally not driven by outlier values and allow for adjustment for age associations with cluster scores. Only clusters which had reproducible statistically significant correlations (p < 0.01) with at least one of the disability measures are represented (Table 3); Correlation p values in the validation cohort were adjusted to account for multiple comparisons (Benjamini and Hochberg, 1995). Only astrocyte cluster 8 and microglia cluster 2 showed reproducible statistically significant correlations with both disability scales and COMRIS-CTD (Table 3 and Figure 2).

Table 3:

Semi-partial Spearman correlation coefficients between cluster scores (after accounting for association with age) and MS disability measures (EDSS and CombiWISE), CNS tissue destruction (COMRIS-CTD) and disease severity (MS-DSS, ARMSS and MSSS). P values for the correlation coefficients in the validation cohort were adjusted using Benjamini Hochberg False Discovery Rate method.

Astrocyte Cluster 8 Microglia Cluster 2
Training Cohort Validation Cohort Training Cohort Validation Cohort
CombiWISE Spearman r 0.30 0.26 0.24 0.27
p value 1.20E-05 1.71E-04 4.00E-04 1.71E-04
EDSS Spearman r 0.29 0.26 0.23 0.24
p value 2.24E-05 3.13 E-04 0.001 4.36E-04
COMRIS-CTD Spearman r 0.38 0.36 0.36 0.36
p value 8.98E-09 1.08E-07 8.51E-08 1.08E-07
MS-DSS Spearman r 0.21 0.19 0.19 0.28
p value 0.003 0.008 0.007 1.34E-04
ARMSS Spearman r 0.27 0.25 0.21 0.24
p value 6.91E-05 0.001 0.002 5.79E-04
MSSS Spearman r 0.19 0.11 0.12 0.12
p value 0.010 0.157 0.097 0.16

Figure 2:

Figure 2:

Scatter plots for Astrocyte Cluster 8 and Microglia Cluster 2; cluster score (adjusted for age) versus MS disability (CombiWISE and EDSS), CNS tissue destruction (COMRIS-CTD) and disease severity (MS-DSS) measures. Both clusters exhibited reproducible statistically significant (p value < 0.01) correlations with clinical measures.

3.5). Correlation Analysis Between Cluster Scores and MS severity

Correlation with disability and CNS tissue destruction identifies processes that develop as MS evolves, but not necessarily processes that drive CNS tissue destruction and disability. Instead, correlation with MS severity, which measures how fast individual patients progress is better suited for identification of potentially pathogenic processes, especially if such MS severity scale can predict future rates of MS progression, as MS-DSS scales can (Weideman et al., 2017a). Therefore, we investigated correlations between CNS cell-enriched biomarker clusters and MS-DSS. We also included assessment of correlations with less sensitive, but more broadly used MS severity scales based on EDSS: ARMSS and MSSS.

Astrocyte cluster 8 and microglia cluster 2, which were elevated during all stages of MS had statistically significant (p < 0.01) correlations with MS-DSS and ARMSS (Table 3 and Figure 2). No cluster correlated with MSSS.

3.6). Analysis of Individual Proteins of Clinically Important Clusters

For astrocyte cluster 8 (MMP7, SERPINA3, GZMA and CLIC1) and microglia cluster 2 (DSG2 and TNFRSF25), individual protein’s SOMAScan RFU values were standardized and accounted for associations with age. Scatter plots for individual proteins were prepared; standardized RFU values (adjusted for age) versus MS disability (EDSS and CombiWISE), CNS tissue destruction (COMRIS-CTD) and disease severity (MS-DSS) measures (Supplementary Figures 2 and 3). Individual proteins of a cluster demonstrated analogous behavior.

3.7). In Vitro Secretion of Cluster Proteins by CNS Cells Under Different Culture Conditions

To aid mechanistic interpretation of our results we asked under which conditions the biomarkers constituting most relevant CNS cell-enriched clusters are produced. We assessed 3 different cell culture conditions: native (control or unstimulated) and two different stressors, pro-inflammatory environment modeled by adding 50% volume/volume supernatants from polyclonally-activated PBMCs, and neuro-degenerative environment modelled by adding mitochondrial toxin rotenone (100 nM). For embryonic stem-cell-derived oligodendrocytes, only native protein secretion was tested due to limited numbers of O4+ cells.

Secretion of individual biomarker under different culture conditions (native, pro-inflammatory stimuli, and mitochondrial stressor) was measured by computing standardized RFU differences between start and end of culture conditions (24h – 0h); then the mean of biomarkers for each cluster under respective culture conditions was computed. Results are presented in Figure 3; only data for two clinically important clusters (astrocyte cluster 8 and microglia cluster 2) is depicted. Secretion of astrocyte cluster 8 was robustly elevated after pro-inflammatory stimuli compared to native secretion. In contrast, secretion of proteins constituting microglial cluster 2 was inhibited by inflammation but increased under mitochondrial stressor.

Figure 3:

Figure 3:

CNS cells were cultured either without any treatment (native) or treated with pro-inflammatory stimuli (stimulated PBMCs supernatant, 50% volume/volume) or mitochondrial stressor (Rotenone, 100 nM). Secretion of individual biomarker under these culture conditions was measured by computing standardized RFU differences between start and end of culture conditions (24h – 0h), and then the mean of each cluster was computed. Only data for clinically important clusters is depicted.

4). DISCUSSION

We consider CSF the body fluid of choice for discovery and development of CNS biomarkers. CNS-derived proteins represent approximately 20% of CSF proteome, and are typically found in higher concentrations in CSF compared to plasma (Reiber, 2003; Thompson, 1995). As CSF is eventually drained to plasma, CNS-derived proteins can be measured in the plasma using highly sensitive assays (Kuhle et al., 2016); however, due to differences in CSF clearance (which is influenced by processes such as cardiac output, sleep, physical activity, and aging) and systemic metabolism, plasma levels explain only 38–60% of the CSF variance (Bielekova and Pranzatelli, 2017). This 40–62% “noise” may require significantly larger cohorts for biomarker discovery and validation. On the other hand, once the simple or complex CNS biomarkers are defined and validated from CSF studies, they may be applicable to large cohorts with matched serum/plasma biological samples (Kuhle et al., 2016). Analyzing clusters of biomarkers is advantageous over analyzing individual biomarkers because it reduces chances of errors in biomarkers identification and provides information regarding probable mechanistic pathways driving disease progression.

In the current study, we have identified CNS cell-enriched protein clusters using in vitro models combined with ex vivo CSF biomarker measurements. The main limitation of this study is the non-physiological reductionist research approach exemplified by simple in vitro culture system(s) of individual CNS cell types. Unfortunately, the environmental influence on CNS cell expression profile is so dynamic (Gosselin et al., 2017), that it is currently impossible to evaluate in-vivo secretome of individual CNS cells. Thus, while using reductionist approach is inevitable, one must be mindful of this limitation and search for creative solutions how to mitigate it. In the future, as more human CNS cell-specific ex-vivo expression data become available (Gosselin et al., 2017), our results may be further expanded or refined using specific cell-enriched expression profiles, to search for secreted proteins. Additionally, we recognize that it is currently not possible to differentiate secretome of human microglia from other cells of myeloid lineage (e.g., blood-derived macrophages and dendritic cells) that were not studied here. Therefore, we will expand our conclusions to encompass myeloid lineage in general. Finally, while this study focused on CNS cells, we recognize that some of the identified proteins are also produced by lymphocytes (e.g., CXCL13 and GZMA) and their production by CNS cells was either unknown or under-appreciated.

We also acknowledge overlap of patients and, consequently, some results between our previously published SOMAscan-based study (Barbour et al., 2017) and current study. In the previous study we used statistical learning to define “molecular diagnostic tests of MS”, represented by CSF biomarker-based model that can differentiate MS patients from subjects with similar phenotypical features (i.e., clinical symptoms or MS-like CNS imaging) or similar pathophysiology (e.g., other inflammatory neurological diseases [OIND]). In analogous manner, we also defined molecular diagnostic test that can differentiate RR-MS from progressive-MS (Barbour et al., 2017). The methodology employed used diagnostic groups (in the training cohort) to search for biomarkers that differ among diagnostic categories irrespective of their cell specificity. Once the molecular tests were defined and validated in the independent cohort(s), we used in-vitro cultures of human immune and CNS cells to interpret the processes/pathways represented by the validated models. Microglial activation and toxic astrogliosis were among processes that help to differentiate RR-MS from progressive-MS, even though statistical learning selected different specific biomarkers reflective of these two intrathecal processes in our previous study (Barbour et al., 2017).

The current project uses opposite methodology of first identifying cell-enriched clusters and then asking whether they differ among diagnostic subgroups. We consider identification of partially overlapping results related to microglial clusters and astroglial clusters highly reassuring.

The most clinically important finding of the current study is the identification of astroglial cluster 8 and microglia cluster 2 as significantly elevated in both progressive- and RRMS patients and reproducibly correlating with measures of MS severity (MS-DSS and ARMSS). These two observations suggest that the aberrant activation of these two glial cell types (and/or other cells of myeloid lineage as described above) represented by these clusters not only happens early in MS disease process but may also contribute to CNS tissue destruction and/or poor clinical recovery. Proteins from both clusters are related to activation of adaptive immune responses. Specifically, TNFRSF25 plays a role in lymphocyte homeostasis by activating NFkB and mediating lymphocyte apoptosis and necroptosis (Bittner et al., 2017). Intriguingly, this receptor was shown to differentially regulate apoptosis of cytotoxic T cells in patients with infectious mononucleosis (IM) than in healthy subjects (Filatova et al., 2016). IM is one of the most potent MS susceptibility factors identified thus far. Thus, it seems plausible that this microglial cluster participates in regulation of cytotoxic CD8+ T cells.

Astrocyte cluster 8 contains metalloproteinase (MMP7), in addition to cytotoxic immune molecule granzyme A (GZMA), chloride intracellular channel 1 (CLIC1) and serpin family A member 3 (SERPINA3), previously linked to neurotoxic reactive astrocyte phenotype induced by pro-inflammatory stimuli (Liddelow et al., 2017a; Padmanabhan et al., 2006; Ritchie et al., 2004). Liddelow et al. have shown that proinflammatory cytokines, mainly interleukin 1 α (IL1α), tumor necrosis factor α (TNFα) and complement component 1, subcomponent q (C1q), released by activated microglia causes induction of neuro-toxic, so called A1astrocytes; they have identified several A1-astrocyte specific markers. By in situ hybridization and immunohistochemistry of post-mortem brain tissue from MS patients they have shown the presence of A1 reactive astrocytes in demyelinating lesions of MS (Liddelow et al., 2017b). We tried to replicate in vitro model of reactive astrocytes by treating primary human astrocytes with supernatants from polyclonally-stimulated PBMCs (unpublished studies). Our model has been validated by checking elevated presence of reactive astrocyte-specific markers, serpin family G member 1 (SERPING1) and complement C3, as described by Liddelow et al. We observed that serpin family A member 3 (SERPINA3; α−1-antichymotrypsin) is a strong marker of A1 reactive astrocytes (almost 62-fold increase in SERPINA3 SOMAscan RFU was observed); and we validated this finding from the microarray data generated by Zamanian et al. deposited in public database (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE35338) (Zamanian et al., 2012). Thus, proteins of astrocyte cluster 8 are highly specific to A1 astrocytes (even though secreted in very low concentration under native condition) and significantly induced under pro-inflammatory stimulation. By demonstrating (and validating) correlation between astrocyte cluster 8 and MS severity, our study expands observation by Liddelow et al. postulating important role of astrocyte-specific processes in MS progression. Accordingly, there is a pressing need for further studies regarding detailed role of toxic astrocytes in MS progression to identify therapeutic targets which may block their neurotoxic potential. Only such interventional clinical trials may determine unequivocally whether neuro-toxic astrocytes play pathogenic role in MS.

Supplementary Material

1

HIGHLIGHTS.

  • Microglial/myeloid cell activation markers are present during all stages of MS.

  • Toxic astrocyte specific markers increase with MS duration.

  • Aberrant activation of glial cells contributes to CNS tissue destruction and enhance MS severity.

5). ACKNOWLEDGEMENTS

The study was supported by the intramural research program of the National Institute of Allergy and Infectious Diseases (NIAID) of the National Institutes of Health (NIH).

ABBREVIATIONS

MS

multiple sclerosis

CNS

central nervous system

CSF

cerebrospinal fluid

SOMAscan

Slow Off-rate Modified DNA Aptamers assay

MMP7

metalloproteinase 7

SERPINA3

serpin family A member 3

GZMA

granzyme A

CLIC1

chloride intracellular channel 1

DSG2

desmoglein 2

TNFRSF25

tumor necrosis factor receptor superfamily member 25

DMTs

disease-modifying treatments

RR-MS

relapsing-remitting MS

SP-MS

secondary-progressive MS

PP-MS

primary-progressive MS

sCD21

soluble cluster of differentiation 21

sCD27

soluble cluster of differentiation 27

EDSS

Expanded Disability Status Scale

SNRS

Scripps Neurological Rating Scale

25FW

25-foot walk

9HPT

nine-hole peg test

CombiWISE

Combinatorial Weight-Adjusted Disability Scale

COMRIS-CTD

Composite MRI scale of CNS tissue destruction

MS-DSS

Multiple Sclerosis Disease Severity Scale

MSSS

Multiple Sclerosis Severity Score

ARMSS

Age Related Multiple Sclerosis Severity

HD

healthy donors

NIND

non-inflammatory neurological disorder

CIS

clinically isolated syndrome

PBMCs

Peripheral Blood Mononuclear Cells

LPS

lipopolysaccharide

ANOVA

Analysis of Variance

SPARC

secreted protein acidic and rich in cysteine

CXCL13

chemokine (C-X-C motif) ligand 13

OIND

other inflammatory neurological diseases

NFkB

nuclear factor kappa-light-chain-enhancer of activated B cells

IM

infectious mononucleosis

IL1α

interleukin 1 α

TNFα

tumor necrosis factor α

C1q

complement component 1, subcomponent q

SERPING1

serpin family G member 1

C3

complement component 3

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

6) REFERENCES

  1. 2018. R: A Language and Environment for Statistical Computing. R foundation for Statitical Computing, Vienna, Austria: URL https://www.R-project.org/. [Google Scholar]
  2. Barbour C, Kosa P, Komori M, Tanigawa M, Masvekar R, Wu T, Johnson K, Douvaras P, Fossati V, Herbst R, Wang Y, Tan K, Greenwood M, Bielekova B, 2017. Molecular-based diagnosis of multiple sclerosis and its progressive stage. Ann Neurol 82(5), 795–812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Benjamini Y, Hochberg Y, 1995. Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. J. R. Statist. Soc B 57(No. 1), 289–300. [Google Scholar]
  4. Bielekova B, Pranzatelli MR, 2017. Promise, Progress, and Pitfalls in the Search for Central Nervous System Biomarkers in Neuroimmunological Diseases: A Role for Cerebrospinal Fluid Immunophenotyping. Semin Pediatr Neurol 24(3), 229–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Bittner S, Knoll G, Ehrenschwender M, 2017. Death receptor 3 mediates necroptotic cell death. Cell Mol Life Sci 74(3), 543–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Choi SR, Howell OW, Carassiti D, Magliozzi R, Gveric D, Muraro PA, Nicholas R, Roncaroli F, Reynolds R, 2012. Meningeal inflammation plays a role in the pathology of primary progressive multiple sclerosis. Brain 135(Pt 10), 2925–2937. [DOI] [PubMed] [Google Scholar]
  7. Douvaras P, Fossati V, 2015. Generation and isolation of oligodendrocyte progenitor cells from human pluripotent stem cells. Nat Protoc 10(8), 1143–1154. [DOI] [PubMed] [Google Scholar]
  8. Filatova EN, Anisenkova EV, Presnyakova NB, Utkin OV, 2016. DR3 regulation of apoptosis of naive T-lymphocytes in children with acute infectious mononucleosis. Acta Microbiol Immunol Hung 63(3), 339–357. [DOI] [PubMed] [Google Scholar]
  9. Frischer JM, Bramow S, Dal-Bianco A, Lucchinetti CF, Rauschka H, Schmidbauer M, Laursen H, Sorensen PS, Lassmann H, 2009. The relation between inflammation and neurodegeneration in multiple sclerosis brains. Brain 132(Pt 5), 1175–1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Gold L, Ayers D, Bertino J, Bock C, Bock A, Brody EN, Carter J, Dalby AB, Eaton BE, Fitzwater T, Flather D, Forbes A, Foreman T, Fowler C, Gawande B, Goss M, Gunn M, Gupta S, Halladay D, Heil J, Heilig J, Hicke B, Husar G, Janjic N, Jarvis T, Jennings S, Katilius E, Keeney TR, Kim N, Koch TH, Kraemer S, Kroiss L, Le N, Levine D, Lindsey W, Lollo B, Mayfield W, Mehan M, Mehler R, Nelson SK, Nelson M, Nieuwlandt D, Nikrad M, Ochsner U, Ostroff RM, Otis M, Parker T, Pietrasiewicz S, Resnicow DI, Rohloff J, Sanders G, Sattin S, Schneider D, Singer B, Stanton M, Sterkel A, Stewart A, Stratford S, Vaught JD, Vrkljan M, Walker JJ, Watrobka M, Waugh S, Weiss A, Wilcox SK, Wolfson A, Wolk SK, Zhang C, Zichi D, 2010. Aptamer-based multiplexed proteomic technology for biomarker discovery. PLoS One 5(12), e15004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Gold L, Walker JJ, Wilcox SK, Williams S, 2012. Advances in human proteomics at high scale with the SOMAscan proteomics platform. N Biotechnol 29(5), 543–549. [DOI] [PubMed] [Google Scholar]
  12. Gosselin D, Skola D, Coufal NG, Holtman IR, Schlachetzki JCM, Sajti E, Jaeger BN, O’Connor C, Fitzpatrick C, Pasillas MP, Pena M, Adair A, Gonda DD, Levy ML, Ransohoff RM, Gage FH, Glass CK, 2017. An environment-dependent transcriptional network specifies human microglia identity. Science 356(6344). [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Hartung HP, Gonsette R, Konig N, Kwiecinski H, Guseo A, Morrissey SP, Krapf H, Zwingers T, Mitoxantrone in Multiple Sclerosis Study, G., 2002. Mitoxantrone in progressive multiple sclerosis: a placebo-controlled, double-blind, randomised, multicentre trial. Lancet 360(9350), 2018–2025. [DOI] [PubMed] [Google Scholar]
  14. Kappos L, Polman C, Pozzilli C, Thompson A, Beckmann K, Dahlke F, European Study Group in Interferon beta-1b in Secondary-Progressive, M.S., 2001. Final analysis of the European multicenter trial on IFNbeta-1b in secondary-progressive MS. Neurology 57(11), 1969–1975. [DOI] [PubMed] [Google Scholar]
  15. Kim S, 2015. ppcor: An R Package for a Fast Calculation to Semi-partial Correlation Coefficients. Commun Stat Appl Methods 22(6), 665–674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Kirk J, Plumb J, Mirakhur M, McQuaid S, 2003. Tight junctional abnormality in multiple sclerosis white matter affects all calibres of vessel and is associated with blood-brain barrier leakage and active demyelination. J Pathol 201(2), 319–327. [DOI] [PubMed] [Google Scholar]
  17. Komori M, Blake A, Greenwood M, Lin YC, Kosa P, Ghazali D, Winokur P, Natrajan M, Wuest SC, Romm E, Panackal AA, Williamson PR, Wu T, Bielekova B, 2015. CSF markers reveal intrathecal inflammation in progressive multiple sclerosis. Ann Neurol 78(1), 3–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Kosa P, Ghazali D, Tanigawa M, Barbour C, Cortese I, Kelley W, Snyder B, Ohayon J, Fenton K, Lehky T, Wu T, Greenwood M, Nair G, Bielekova B, 2016. Development of a Sensitive Outcome for Economical Drug Screening for Progressive Multiple Sclerosis Treatment. Front Neurol 7, 131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Kosa P, Komori M, Waters R, Wu T, Cortese I, Ohayon J, Fenton K, Cherup J, Gedeon T, Bielekova B, 2015. Novel composite MRI scale correlates highly with disability in multiple sclerosis patients. Mult Scler Relat Disord 4(6), 526–535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Kraemer S, Vaught JD, Bock C, Gold L, Katilius E, Keeney TR, Kim N, Saccomano NA, Wilcox SK, Zichi D, Sanders GM, 2011. From SOMAmer-based biomarker discovery to diagnostic and clinical applications: a SOMAmer-based, streamlined multiplex proteomic assay. PLoS One 6(10), e26332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Kroksveen AC, Jaffe JD, Aasebo E, Barsnes H, Bjorlykke Y, Franciotta D, Keshishian H, Myhr KM, Opsahl JA, van Pesch V, Teunissen CE, Torkildsen O, Ulvik RJ, Vethe H, Carr SA, Berven FS, 2015. Quantitative proteomics suggests decrease in the secretogranin-1 cerebrospinal fluid levels during the disease course of multiple sclerosis. Proteomics 15(19), 3361–3369. [DOI] [PubMed] [Google Scholar]
  22. Kuhle J, Barro C, Disanto G, Mathias A, Soneson C, Bonnier G, Yaldizli O, Regeniter A, Derfuss T, Canales M, Schluep M, Du Pasquier R, Krueger G, Granziera C, 2016. Serum neurofilament light chain in early relapsing remitting MS is increased and correlates with CSF levels and with MRI measures of disease severity. Mult Scler 22(12), 1550–1559. [DOI] [PubMed] [Google Scholar]
  23. Kurtzke JF, 1983. Rating neurologic impairment in multiple sclerosis: an expanded disability status scale (EDSS). Neurology 33(11), 1444–1452. [DOI] [PubMed] [Google Scholar]
  24. Kutzelnigg A, Lucchinetti CF, Stadelmann C, Bruck W, Rauschka H, Bergmann M, Schmidbauer M, Parisi JE, Lassmann H, 2005. Cortical demyelination and diffuse white matter injury in multiple sclerosis. Brain 128(Pt 11), 2705–2712. [DOI] [PubMed] [Google Scholar]
  25. Lassmann H, 2016. Demyelination and neurodegeneration in multiple sclerosis: The role of hypoxia. Ann Neurol 79(4), 520–521. [DOI] [PubMed] [Google Scholar]
  26. Lassmann H, van Horssen J, 2011. The molecular basis of neurodegeneration in multiple sclerosis. FEBS Lett 585(23), 3715–3723. [DOI] [PubMed] [Google Scholar]
  27. Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, Bennett ML, Munch AE, Chung WS, Peterson TC, Wilton DK, Frouin A, Napier BA, Panicker N, Kumar M, Buckwalter MS, Rowitch DH, Dawson VL, Dawson TM, Stevens B, Barres BA, 2017a. Neurotoxic reactive astrocytes are induced by activated microglia. Nature [DOI] [PMC free article] [PubMed]
  28. Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, Bennett ML, Munch AE, Chung WS, Peterson TC, Wilton DK, Frouin A, Napier BA, Panicker N, Kumar M, Buckwalter MS, Rowitch DH, Dawson VL, Dawson TM, Stevens B, Barres BA, 2017b. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541(7638), 481–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Magliozzi R, Howell O, Vora A, Serafini B, Nicholas R, Puopolo M, Reynolds R, Aloisi F, 2007. Meningeal B-cell follicles in secondary progressive multiple sclerosis associate with early onset of disease and severe cortical pathology. Brain 130(Pt 4), 1089–1104. [DOI] [PubMed] [Google Scholar]
  30. Mahad D, Lassmann H, Turnbull D, 2008. Review: Mitochondria and disease progression in multiple sclerosis. Neuropathol Appl Neurobiol 34(6), 577–589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Manouchehrinia A, Westerlind H, Kingwell E, Zhu F, Carruthers R, Ramanujam R, Ban M, Glaser A, Sawcer S, Tremlett H, Hillert J, 2017. Age Related Multiple Sclerosis Severity Score: Disability ranked by age. Mult Scler 23(14), 1938–1946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. McMahon JM, McQuaid S, Reynolds R, FitzGerald UF, 2012. Increased expression of ER stress- and hypoxia-associated molecules in grey matter lesions in multiple sclerosis. Mult Scler 18(10), 1437–1447. [DOI] [PubMed] [Google Scholar]
  33. Mohajeri M, Sadeghizadeh M, Najafi F, Javan M, 2015. Polymerized nano-curcumin attenuates neurological symptoms in EAE model of multiple sclerosis through down regulation of inflammatory and oxidative processes and enhancing neuroprotection and myelin repair. Neuropharmacology 99, 156–167. [DOI] [PubMed] [Google Scholar]
  34. Montalban X, Hauser SL, Kappos L, Arnold DL, Bar-Or A, Comi G, de Seze J, Giovannoni G, Hartung HP, Hemmer B, Lublin F, Rammohan KW, Selmaj K, Traboulsee A, Sauter A, Masterman D, Fontoura P, Belachew S, Garren H, Mairon N, Chin P, Wolinsky JS, Investigators OC, 2016. Ocrelizumab versus Placebo in Primary Progressive Multiple Sclerosis. N Engl J Med [DOI] [PubMed]
  35. Noble PB, Cutts JH, 1967. Separation of blood leukocytes by Ficoll gradient. Can Vet J 8(5), 110–111. [PMC free article] [PubMed] [Google Scholar]
  36. Padmanabhan J, Levy M, Dickson DW, Potter H, 2006. Alpha1-antichymotrypsin, an inflammatory protein overexpressed in Alzheimer’s disease brain, induces tau phosphorylation in neurons. Brain 129(Pt 11), 3020–3034. [DOI] [PubMed] [Google Scholar]
  37. Reiber H, 2003. Proteins in cerebrospinal fluid and blood: barriers, CSF flow rate and source-related dynamics. Restor Neurol Neurosci 21(3–4), 79–96. [PubMed] [Google Scholar]
  38. Revesz T, Kidd D, Thompson AJ, Barnard RO, McDonald WI, 1994. A comparison of the pathology of primary and secondary progressive multiple sclerosis. Brain 117 ( Pt 4), 759–765. [DOI] [PubMed] [Google Scholar]
  39. Reynolds R, Roncaroli F, Nicholas R, Radotra B, Gveric D, Howell O, 2011. The neuropathological basis of clinical progression in multiple sclerosis. Acta Neuropathol. Ritchie, A., Morgan, K., Kalsheker, N., 2004. Allele-specific overexpression in astrocytes of an Alzheimer’s disease associated alpha-1-antichymotrypsin promoter polymorphism. Brain Res Mol Brain Res 131(1–2), 88–92. [DOI] [PubMed] [Google Scholar]
  40. Rohloff JC, Gelinas AD, Jarvis TC, Ochsner UA, Schneider DJ, Gold L, Janjic N, 2014. Nucleic Acid Ligands With Protein-like Side Chains: Modified Aptamers and Their Use as Diagnostic and Therapeutic Agents. Mol Ther Nucleic Acids 3, e201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Roxburgh RH, Seaman SR, Masterman T, Hensiek AE, Sawcer SJ, Vukusic S, Achiti I, Confavreux C, Coustans M, le Page E, Edan G, McDonnell GV, Hawkins S, Trojano M, Liguori M, Cocco E, Marrosu MG, Tesser F, Leone MA, Weber A, Zipp F, Miterski B, Epplen JT, Oturai A, Sorensen PS, Celius EG, Lara NT, Montalban X, Villoslada P, Silva AM, Marta M, Leite I, Dubois B, Rubio J, Butzkueven H, Kilpatrick T, Mycko MP, Selmaj KW, Rio ME, Sa M, Salemi G, Savettieri G, Hillert J, Compston DA, 2005. Multiple Sclerosis Severity Score: using disability and disease duration to rate disease severity. Neurology 64(7), 1144–1151. [DOI] [PubMed] [Google Scholar]
  42. Sipe JC, Knobler RL, Braheny SL, Rice GP, Panitch HS, Oldstone MB, 1984. A neurologic rating scale (NRS) for use in multiple sclerosis. Neurology 34(10), 1368–1372. [DOI] [PubMed] [Google Scholar]
  43. Stevens J, 2002. Applied multivariate statistics for the social sciences, 4th ed. Lawrence Erlbaum Associates, Mahwah, N.J. [Google Scholar]
  44. Stys PK, Zamponi GW, van Minnen J, Geurts JJ, 2012. Will the real multiple sclerosis please stand up? Nat Rev Neurosci 13(7), 507–514. [DOI] [PubMed] [Google Scholar]
  45. Thompson EJ, 1995. Cerebrospinal fluid. J Neurol Neurosurg Psychiatry 59(4), 349–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Weideman AM, Barbour C, Tapia-Maltos MA, Tran T, Jackson K, Kosa P, Komori M, Wichman A, Johnson K, Greenwood M, Bielekova B, 2017a. New Multiple Sclerosis Disease Severity Scale Predicts Future Accumulation of Disability. Front Neurol 8, 598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Weideman AM, Tapia-Maltos MA, Johnson K, Greenwood M, Bielekova B, 2017b. Meta-analysis of the Age-Dependent Efficacy of Multiple Sclerosis Treatments. Front Neurol 8, 577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Weksler BB, Subileau EA, Perriere N, Charneau P, Holloway K, Leveque M, Tricoire-Leignel H, Nicotra A, Bourdoulous S, Turowski P, Male DK, Roux F, Greenwood J, Romero IA, Couraud PO, 2005. Blood-brain barrier-specific properties of a human adult brain endothelial cell line. FASEB J 19(13), 1872–1874. [DOI] [PubMed] [Google Scholar]
  49. Whittaker J, 1990. Graphical Models in Applied Multivariate Statistics John Wiley & Sons, Chichester. [Google Scholar]
  50. Zamanian JL, Xu L, Foo LC, Nouri N, Zhou L, Giffard RG, Barres BA, 2012. Genomic analysis of reactive astrogliosis. J Neurosci 32(18), 6391–6410. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

RESOURCES