Skip to main content

Investigation of sex-specific effects of apolipoprotein E on severity of EAE and MS

Abstract

Background

Despite pleiotropic immunomodulatory effects of apolipoprotein E (apoE) in vitro, its effects on the clinical course of experimental autoimmune encephalomyelitis (EAE) and multiple sclerosis (MS) are still controversial. As sex hormones modify immunomodulatory apoE functions, they may explain contentious findings. This study aimed to investigate sex-specific effects of apoE on disease course of EAE and MS.

Methods

MOG35-55 induced EAE in female and male apoE-deficient mice was assessed clinically and histopathologically. apoE expression was investigated by qPCR. The association of the MS severity score (MSSS) and APOE rs429358 and rs7412 was assessed across 3237 MS patients using linear regression analyses.

Results

EAE disease course was slightly attenuated in male apoE-deficient (apoE −/−) mice compared to wildtype mice (cumulative median score: apoE −/− = 2 [IQR 0.0–4.5]; wildtype = 4 [IQR 1.0–5.0]; n = 10 each group, p = 0.0002). In contrast, EAE was more severe in female apoE −/− mice compared to wildtype mice (cumulative median score: apoE −/− = 3 [IQR 2.0–4.5]; wildtype = 3 [IQR 0.0–4.0]; n = 10, p = 0.003). In wildtype animals, apoE expression during the chronic EAE phase was increased in both females and males (in comparison to naïve animals; p < 0.001). However, in MS, we did not observe a significant association between MSSS and rs429358 or rs7412, neither in the overall analyses nor upon stratification for sex.

Conclusions

apoE exerts moderate sex-specific effects on EAE severity. However, the results in the apoE knock-out model are not comparable to effects of polymorphic variants in the human APOE gene, thus pinpointing the challenge of translating findings from the EAE model to the human disease.

Background

Apolipoprotein E (apoE) exerts pleiotropic biological functions, including effects on lipoprotein metabolism as well as on the innate and adaptive immune system. Potential mechanisms underlying the immunomodulatory properties of apoE involve enhanced anti-inflammatory macrophage phenotype, decreased activation of NF-kB and STAT1 [1], and downregulation of TH-1 and TH-17 responses via suppression of pro-inflammatory cytokines secreted by macrophages [2]. apoE is expressed in the CNS and is produced by antigen-presenting cells (dendritic cells, macrophages). These observations have led to the investigation of apoE in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE; reviewed by [3]). In this context, controversial results have been reported for apoE in EAE including both beneficial as well as aggravating effects on disease severity and progression in apoE knock-out mice [4, 5]. In parallel, two APOE polymorphisms, i.e., rs429358 (ε4, Cys130Arg) and rs7412 (ε2, Arg176Cys), which represent established risk variants in Alzheimer’s disease [6], have been assessed extensively for their role in MS. A recent study compiling data on nearly 30,000 subjects showed that these polymorphisms do not influence MS susceptibility [7]. However, their role in disease progression still remains ambiguous, which at least in part pertains to the fact that the majority of studies have assessed rather small, i.e., underpowered datasets (for an overview see e.g., [8]). Divergent findings may also be due to confounders or effect modifiers such as sex, age, or patient subgroups. Along these lines, a comparatively small study testing 221 patients suggested that the association between APOE and MS severity was limited to women [9]; however, this has not been described in other studies [8]. Thus, in the current study, we comprehensively assessed the role of apolipoprotein E on disease severity of EAE as well as MS by taking into consideration potential sex-specific effects of APOE genotypes.

Methods

Mice, experimental autoimmune encephalomyelitis, histopathology, and quantitative real-time PCR analyses

Animal experiments were approved by the North-Rhine-Westphalia authorities for animal experimentation (AZ 84–02.04.2011.A251). Wildtype (wt) C57BL/6 (Harlan, Germany) and apoE −/− mice (University of Duisburg-Essen, Germany) were backcrossed to generate littermates. Chronic EAE was induced in male and female 9–11-week-old mice using 100 μg myelin-oligodendrocyte glycoprotein peptide (MOG35-55) (Charité Berlin, Germany) emulsified in complete Freund’s adjuvant (CFA) containing 100 μg Mycobacterium tuberculosis H37RA (Difco Laboratories, Augsburg, Germany) with pertussis toxin injections (PTX, 100 ng intraperitoneally) (LuBio Science, Luzern, Switzerland) on day 0 (d0) and d2 post-immunization (p.i.). Two independent experiments, each including both genotypes and sexes, were performed. As controls, only littermate animals were used. Clinical EAE signs were evaluated daily using a 10-grade score [10] by an experimenter blinded to the genotype. Concentration of plasma neurofilament heavy chain (NfH) was quantified by ELISA as described previously [11, 12]. The amount of NfH for each animal was calculated as the difference between NfH concentration at d26 after immunization and at baseline (i.e., before induction of EAE). For histopathology, mice were perfused during the chronic disease phase of EAE (d26 or d35), and immunohistochemistry was performed on cryosections of lumbar spinal cord tissue for T cells (rat-α-human CD3, 1:100; AbD Serotec, Düsseldorf, Germany) and macrophages (rat-α-mouse Mac3, 1:100; BD-Pharmingen, Heidelberg, Germany) [13]. Demyelination was assessed using FluoroMyelin™ Red Fluorescent myelin stain (1:300) according to manufacturer’s protocol (Life Technologies, Karlsruhe, Germany) with DAPI counterstaining of nuclei (Southern Biotech, Birmingham, USA). Fluorescent images were captured using an inverted fluorescence microscope (BX51, Olympus), and the percentage of demyelinated area was determined by ImageJ [4]. Data are presented as median [interquartile range [IQR], i.e., 25–75. percentile] or mean ± standard error (SEM). To determine differences in clinical course of EAE, Mann-Whitney U test was performed for cumulative median scores (from onset of disease (d8) until the end of observation (d35), statistical significance is graphically indicated as **p < 0.01, ***p < 0.001) as well as median score for individual time points (statistical significance is graphically indicated as #p < 0.05). Effects of apoE genotypes on NfH were analyzed using Mann-Whitney U test and on histological parameters (amount of T cells, macrophages, demyelination, axonal density) using Student’s t test.

Quantitative real-time PCR (q-rtPCR) for relative apoE expression in the spinal cord of male and female wt mice was performed during the chronic disease phase (d35 p.i.). Total RNA was isolated using TRIZOL followed by the RNAeasy Mini Kit (Quiagen, Hilden, Germany) and transcribed to cDNA according to the manufacturer’s protocol (DNAse1 (Invitrogen, Karlsruhe, Germany); anchored Oligo-dt (Thermo Fischer, Schwerte, Germany); dNTPs (Invitrogen); Superscript II (Invitrogen)). Q-rtPCR was performed on an ABI real-time PCR system (Applied Biosystems, Darmstadt, Germany) using PerfeCTa FastMixII master mix (Quanta Bioscience, Gaithersburg, USA) (primer: Mm01307193_g1, Applied Biosystems) normalized to the housekeeping gene β-actin (primer: Mm00607939_s1 Actb, Applied Biosystems) using the ∆∆ct method. Differences of sex and EAE on apoE expression were calculated using a one-way ANOVA followed by Bonferroni’s multiple comparison test.

Results were calculated using GraphPad Prism 6 (GraphPad Software, USA). In all experiments, a p value of <0.05 was defined as statistically significant, p values ≥0.05 as non-significant (n.s.).

Human subjects and genotyping

All samples were collected after informed written consent and appropriate ethical approval at the respective sites. The current study included 2193 MS cases (70.6 % women) from Germany and 1044 patients (71.0 % women) from France, for whom APOE genotypes had been generated previously (see [7] for details) and for whom information on the expanded disability status scale (EDSS) [14], disease duration at the timepoint of EDSS assessment, and age at onset (AAO) was available. In case of multiple EDSS measurements, only the most recent one was used. Based on the available data on the EDSS and disease duration, the multiple sclerosis severity score (MSSS) was calculated [15]. See [7], Additional file 1: Table S1 and Additional file 1: Figure S3 for demographic details.

Power calculation and genetic association analysis

Power estimates for the human association analyses were calculated using the genetic power calculator [16, 17] assuming a minor allele frequency of 0.15 and 0.07 as reported in NCBI’s dbSNP [18], a type I error rate of 0.05, and an additive quantitative trait locus (QTL) model. For both polymorphisms, our study had excellent (>90 %) power to detect additive QTL variance of 0.5 %. Linear regression analyses using an additive and a recessive model were performed in PLINK v1.07 [19, 20] including all subjects and adjusting for center of recruitment, AAO, and sex. Additional analyses were performed after stratification for sex while adjusting for center and AAO. Empirical p values were obtained after 10,000 rounds of permutation. All reported p values are two-tailed.

Results

apoE deficiency ameliorates EAE course in male but not female mice

apoE deficiency demonstrated a moderate sex-specific effect on the clinical EAE course. In male animals, cumulative disease severity was attenuated in apoE −/− mice compared to wt mice (cumulative median score: male apoE −/− = 2 [IQR 0.0–4.5], n = 10, male apoE +/+ = 4 [IQR 1.0–5.0], n = 10; p = 0.0002; Fig. 1a). Considering median clinical scores for individual time points, male mice showed a significant difference between genotypes on d21 and d22 p.i. (p < 0.05). Following this line, body weight was higher in male apoE −/− mice (cumulative mean weight 28.1 ± 0.9 g) than in male wt animals (cumulative mean weight, 26.2 ± 0.8 g; p < 0.001; Additional file 1: Figure S1a). In contrast, in the female group, EAE was more severe in apoE −/− mice compared to wt controls (cumulative median score: female apoE −/− = 3 [IQR 2.0–4.5], n = 10, female apoE +/+ = 3 [IQR 0.0–4.0], n = 10; p = 0.003; Fig. 1b and cumulative mean weight: female wt = 20.5 ± 0.7 g; female apoE −/− = 20.5 ± 0.8 g; p = n.s.; Additional file 1: Figure S1b). Differences in EAE incidence supported the sex-specific effect of the apoE genotype on disease manifestation (incidence male apoE −/− = 7/10, male apoE +/+ = 10/10, female apoE −/− = 10/10, female apoE +/+ = 9/10, p = n.s., Fisher’s exact test). These results confirmed our initial observations in pilot studies with non-littermate control animals (data not shown).

Fig. 1
figure 1

Clinical disease course of active MOG35-55 induced EAE in C57Bl/6 wildtype (apoE +/+) and apoE-deficient (apoE −/−) mice. a In male (m) mice, the absence of apoE leads to an attenuated disease severity. b In contrast, in females (f), apoE deficiency results in a worse disease course. Data were pooled from two independent experiments. Clinical disease course was assessed using a 10-grade scale and depicted as mean ± SEM. Statistical analyses: median scores analyzed with Mann-Whitney U test from onset of disease (d8) until the end of observation (d35) (***p < 0.001, **p < 0.01) and for individual time points (#p < 0.05)

In line with the clinical data, neurofilament heavy chain (NfH) concentrations in the plasma, which represent a marker for axonal damage [11, 21, 22] revealed a significant difference between male wt and apoE-deficient mice (p = 0.0286) but not between female mice (p = 0.8; Fig. 2). Significantly increased axonal degeneration in spinal cord sections of male wt animals compared to male apoE-deficient mice (d35) was further corroborated by silver impregnation (relative axonal density: male apoE −/− = 10.2 ± 3.2, n = 6, male apoE +/+ = 3.2 ± 0.8, n = 5; p = 0.001).

Fig. 2
figure 2

Neurofilament heavy chain (NfH) concentration in blood plasma of C57Bl/6 wildtype mice (apoE +/+) and apoE-deficient (apoE −/−) mice after a MOG35-55-induced EAE indicates the extent of axonal damage. In the chronic phase (d26), male apoE +/+ mice show a significantly increased NfH concentration compared to male apoE −/− mice. NfH concentration was calculated as difference (Δ) between d26 and baseline (i.e., before EAE-induction). Statistical analyses: *p < 0.05 by Mann-Whitney U test

Spinal cord immunohistopathology (pooled data from d26 and d35) did not show statistically significant differences of apoE −/− mice compared to wt mice within the two sex strata; however, observations tended to be consistent with the clinical observations. Specifically, male apoE −/− mice showed slightly lower macrophage (−29 %, p = 0.4, Additional file 1: Figure S2a) and T-cell infiltration (−46 %, p = 0.3; Additional file 1: Figure S2b) compared to male wt mice. Likewise, female apoE −/− mice showed 47 % more infiltrating macrophages (p = 0.06; Additional file 1: Figure S2a) and 34 % more T cells (p = 0.3; Additional file 1: Figure S2b) than the wt group. Furthermore, male apoE −/− mice showed a non-significant decrease in demyelination compared to male controls (wt = 8.6 % ± 4.8, n = 5, apoE −/− = 5.3 % ± 5.2; n = 5; p = 0.3), and a non-significant effect pointing into the opposite direction was observed for females (wt = 2.0 % ± 1.5, n = 8, apoE −/− = 4.3 % ± 3.4; n = 5; p = 0.1; Additional file 1: Figure S2c).

During the chronic EAE phase, apoE expression in the spinal cord increased in wt animals in comparison to untreated healthy controls in both strata (females: 13-fold increase; males: 11-fold increase, one-way ANOVA, p < 0.001; Fig. 3).

Fig. 3
figure 3

Relative apoE expression in the spinal cord at different time points of a MOG35-55-induced EAE in C57Bl/6 wildtype mice. Quantitative real-time PCR analyses show an increased apoE expression in the chronic phase (d35) of EAE in both females and males compared to untreated animals (BL). Statistical analyses: ***p < 0.001; one-way ANOVA (post test: Bonferroni’s multiple comparison test)

Association analyses of MSSS and APOE polymorphisms do not show statistically significant results

Linear regression analyses of MSSS and APOE rs7412 and rs429358 in the overall analyses across 3237 MS patients did not reveal statistically significant (p < 0.05) association with MS severity after 10,000 rounds of permutation. This result did not change after stratification for sex (Table 1, Additional file 1: Figure S4). While the minor (C) allele of rs429358 tended to be associated with a higher MSSS across all patients assuming a recessive model (β = 0.841, p rec = 0.0140) and in the female stratum assuming both recessive (β = 0.971, p rec = 0.0170) as well as additive models (β = 0.221, p add = 0.0464), significance of neither of these results survived after 10,000 rounds of permutation (Table 1).

Table 1 Association analyses of the MS severity score and APOE polymorphisms rs429358 and rs7412

Discussion

Results of this study indicate that the absence of apoE slightly attenuates EAE in male mice but at the same time aggravates disease course in female animals. In line with this observation, decreased NfH concentration in male apoE-deficient mice in comparison to wt mice suggests an attenuation of axonal damage in male mice lacking apoE. Increased apoE expression in the spinal cord of female and male wt mice in the chronic disease phase may indicate an influence of apoE on disease progression during EAE.

In contrast to the results in the rodent model, we did not detect a robust association between MSSS and APOE rs7412 or rs429358 in over 3200 patients despite excellent (>90 %) power to observe even moderate changes in the MSSS. This suggests that rs7412 and rs429358 do not have a notable influence on MS severity.

Studies that investigated the role of apoE deficiency in EAE have yielded inconsistent, in parts, and even contradictory results [2, 4, 5, 23]. Discrepancies may be due to methodological differences (e.g., the immunization protocol) or due to other modifying factors that have not been investigated in the respective studies. In this context, one potential influencing factor that was not controlled for in previous studies is the sex distribution of the tested animals.

apoE is expressed in the CNS in resident immune cells and has been implicated in different immunoregulatory functions [1, 2, 23]. For instance, in a recent study, milder disease in apoE-deficient mice was associated with a reduction of dendritic cells (DCs), which—in turn—can be modulated by sex hormones, i.e., estrogens and primarily E2 [24, 25]. Other studies have reported that apoE modulates macrophages toward an anti-inflammatory phenotype [1] and suppresses microglial activation [26, 27]. The activity of these cells can be modulated by the exposition of estrogen and testosterone (reviewed in [28]). Androgene-receptors (AR) are expressed on immune cells [29]; therefore, especially, androgens may have immunomodulatory or even immunosuppressive effects [28]. A direct interaction of apoE with AR has also been described [3032]. Thus, immune functions appear to be influenced by androgens via AR and may be modulated by apoE. Although we did not investigate mechanistic pathways, the previously described interactions between immune functions and sex hormones may account for some of the sex-specific differences observed in our study that may additionally be influenced by apoE.

While our human data do not reveal sex-specific association of APOE genotypes and MS severity, the association of APOE genotypes with Alzheimer’s disease (AD) has been described to be modulated by sex and ethnicity. Whereas APOE2 and APOE3 seem to be protective across ethnic groups, APOE4 increases AD risk [6]. The latter effect appears to be pronounced in women [33].

The lack of association of tested APOE polymorphisms with MS severity is in line with the results of most previous publications (for an overview see [8]), including a large pooled re-analysis of previously published datasets on 3518 patients [34] that are independent from those analyzed here. Overall, the authors of the latter study did not find compelling evidence for an association of APOE and MSSS either. While they observed a higher MSSS in male homozygote carriers of the APOE e4 allele when compared to all other groups (p = 0.004), this finding did not withstand multiple comparison corrections [34]. In light of the fact that the two largest, independent, and well-powered studies on rs7412 and rs42935 did not produce robust results, it appears most likely that rs7412 and rs429358 in APOE do not play a substantial role in MS severity as measured by the MSSS. However, several aspects need to be considered upon interpretation of our association results: We have tested two APOE variants, namely two non-synonymous polymorphisms that represent the most important contributors to Alzheimer’s disease risk [6] and that have been extensively characterized functionally. However, even homozygosity at either of these polymorphic sites does not fully mimic the rodent apoE knock-out model [35]. Therefore, the lack of robust genetic effects in humans does not necessarily contradict the results obtained in the apoE −/− mouse model. However, the translation of findings from experimental models and especially in the context of EAE to the human situation has repeatedly failed as only certain facets of the human disease can be modeled [36]. Thus, we cannot exclude that the effects of apoE observed in the rodent EAE model in this study are of lesser or no relevance for the human disease. In addition, we have only assessed the aforementioned two non-synonymous polymorphisms in the APOE region. Thus, we cannot exclude the presence of other variants in the APOE locus with a possible effect on MS severity, although a recent genome-wide association study (which did not assess those two variants directly due to technical reasons (see [7] for explanation)), did not observe evidence for an association of MSSS and other genetic variants in the APOE region [37]. Another consideration extends to the fact that genetic association analyses of MS severity have overall only yielded rather limited success [38]; one explanation, which could also affect the MSSS association analysis results presented here, is the lack of more appropriate clinical and paraclinical classification schemes to better represent disease severity and progression. In addition, other variables, e.g., information on treatment regimes, may represent confounders in the APOE association analysis that could not be accounted for in our and previous analyses (e.g., [34]).

Conclusions

In conclusion, our study shows a moderate sex-specific influence of apoE on EAE severity indicating a complex interaction between apoE, sex, and inflammatory processes at least in the animal model. We did not observe robust sex-specific effects of APOE polymorphisms on MS severity, which may be explained by several factors including difficulties in comparing rodent apoE-deficient animals and polymorphic changes in the human APOE gene. Further characterization of apoE and its potential sex-specific influences on inflammation may lead to novel insights into disease-modifying mechanisms. Yet, our study highlights difficulties of direct translation of experimental findings in mice to the human situation.

References

  1. Baitsch D, Bock HH, Engel T, Telgmann R, Muller-Tidow C, Varga G, et al. Apolipoprotein E induces antiinflammatory phenotype in macrophages. Arterioscler Thromb Vasc Biol. 2011;31:1160–8.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  2. Wei J, Zheng M, Liang P, Wei Y, Yin X, Tang Y, et al. Apolipoprotein E and its mimetic peptide suppress Th1 and Th17 responses in experimental autoimmune encephalomyelitis. Neurobiol Dis. 2013;56:59–65.

    Article  CAS  PubMed  Google Scholar 

  3. Zhang HL, Wu J, Zhu J. The immune-modulatory role of apolipoprotein E with emphasis on multiple sclerosis and experimental autoimmune encephalomyelitis. Clin Dev Immunol. 2010;2010:186813.

    PubMed Central  PubMed  Google Scholar 

  4. Dayger CA, Rosenberg JS, Winkler C, Foster S, Witkowski E, Benice TS, et al. Paradoxical effects of apolipoprotein E on cognitive function and clinical progression in mice with experimental autoimmune encephalomyelitis. Pharmacol Biochem Behav. 2013;103:860–8.

    Article  CAS  PubMed  Google Scholar 

  5. Karussis D, Michaelson DM, Grigoriadis N, Korezyn AD, Mizrachi-Koll R, Chapman S, et al. Lack of apolipoprotein-E exacerbates experimental allergic encephalomyelitis. Mult Scler. 2003;9:476–80.

    Article  CAS  PubMed  Google Scholar 

  6. Farrer LA, Cupples LA, Haines JL, Hyman B, Kukull WA, Mayeux R, et al. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consortium. JAMA. 1997;278:1349–56.

    Article  CAS  PubMed  Google Scholar 

  7. Lill CM, Liu T, Schjeide BM, Roehr JT, Akkad DA, Damotte V, et al. Closing the case of APOE in multiple sclerosis: no association with disease risk in over 29 000 subjects. J Med Genet. 2012;49:558–62.

    Article  CAS  PubMed  Google Scholar 

  8. van der Walt A, Stankovich J, Bahlo M, Taylor BV, van der Mei IA, Foote SJ, et al. Apolipoprotein genotype does not influence MS severity, cognition, or brain atrophy. Neurology. 2009;73:1018–25.

    Article  PubMed  Google Scholar 

  9. Kantarci OH, Hebrink DD, Achenbach SJ, Pittock SJ, Altintas A, Schaefer-Klein JL, et al. Association of APOE polymorphisms with disease severity in MS is limited to women. Neurology. 2004;62:811–4.

    Article  CAS  PubMed  Google Scholar 

  10. Linker RA, Maurer M, Gaupp S, Martini R, Holtmann B, Giess R, et al. CNTF is a major protective factor in demyelinating CNS disease: a neurotrophic cytokine as modulator in neuroinflammation. Nat Med. 2002;8:620–4.

    Article  CAS  PubMed  Google Scholar 

  11. Gresle MM, Shaw G, Jarrott B, Alexandrou EN, Friedhuber A, Kilpatrick TJ, et al. Validation of a novel biomarker for acute axonal injury in experimental autoimmune encephalomyelitis. J Neurosci Res. 2008;86:3548–55.

    Article  CAS  PubMed  Google Scholar 

  12. Petzold A, Keir G, Green AJ, Giovannoni G, Thompson EJ. A specific ELISA for measuring neurofilament heavy chain phosphoforms. J Immunol Methods. 2003;278:179–90.

    Article  CAS  PubMed  Google Scholar 

  13. Linker RA, Lee DH, Ryan S, van Dam AM, Conrad R, Bista P, et al. Fumaric acid esters exert neuroprotective effects in neuroinflammation via activation of the Nrf2 antioxidant pathway. Brain : a journal of neurology. 2011;134:678–92.

    Article  Google Scholar 

  14. Kurtzke JF. Rating neurologic impairment in multiple sclerosis: an expanded disability status scale (EDSS). Neurology. 1983;33:1444–52.

    Article  CAS  PubMed  Google Scholar 

  15. Roxburgh RH, Seaman SR, Masterman T, Hensiek AE, Sawcer SJ, Vukusic S, et al. Multiple sclerosis severity score: using disability and disease duration to rate disease severity. Neurology. 2005;64:1144–51.

    Article  CAS  PubMed  Google Scholar 

  16. Genetic power calculator. [http://pngu.mgh.harvard.edu/~purcell/gpc/qtlassoc.html], last accessed 15-Aug-2015.

  17. Purcell S, Cherny SS, Sham PC. Genetic power calculator: design of linkage and association genetic mapping studies of complex traits. Bioinformatics. 2003;19:149–50.

    Article  CAS  PubMed  Google Scholar 

  18. National Center for Biotechnology Information: dbSNP. [http://www.ncbi.nlm.nih.gov/snp/], last accessed 15-Aug-2015.

  19. Purcell S: PLINKv1.07. [http://pngu.mgh.harvard.edu/purcell/plink/], last accessed 15-Aug-2015.

  20. Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MA, Bender D, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81:559–75.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  21. Petzold A. CSF biomarkers for improved prognostic accuracy in acute CNS disease. Neurol Res. 2007;29:691–708.

    Article  CAS  PubMed  Google Scholar 

  22. Petzold A, Brassat D, Mas P, Rejdak K, Keir G, Giovannoni G, et al. Treatment response in relation to inflammatory and axonal surrogate marker in multiple sclerosis. Mult Scler. 2004;10:281–3.

    Article  CAS  PubMed  Google Scholar 

  23. Shin S, Walz KA, Archambault AS, Sim J, Bollman BP, Koenigsknecht-Talboo J, et al. Apolipoprotein E mediation of neuro-inflammation in a murine model of multiple sclerosis. J Neuroimmunol. 2014;271:8–17.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  24. Robinson DP, Hall OJ, Nilles TL, Bream JH, Klein SL. 17beta-estradiol protects females against influenza by recruiting neutrophils and increasing virus-specific CD8 T cell responses in the lungs. J Virol. 2014;88:4711–20.

    Article  PubMed Central  PubMed  Google Scholar 

  25. Straub RH. The complex role of estrogens in inflammation. Endocr Rev. 2007;28:521–74.

    Article  CAS  PubMed  Google Scholar 

  26. Christensen DJ, Ohkubo N, Oddo J, Van Kanegan MJ, Neil J, Li F, et al. Apolipoprotein E and peptide mimetics modulate inflammation by binding the SET protein and activating protein phosphatase 2A. J Immunol. 2011;186:2535–42.

    Article  CAS  PubMed  Google Scholar 

  27. Laskowitz DT, Thekdi AD, Thekdi SD, Han SK, Myers JK, Pizzo SV, et al. Downregulation of microglial activation by apolipoprotein E and apoE-mimetic peptides. Exp Neurol. 2001;167:74–85.

    Article  CAS  PubMed  Google Scholar 

  28. Trigunaite A, Dimo J, Jorgensen TN. Suppressive effects of androgens on the immune system. Cell Immunol. 2015;294:87–94.

    Article  CAS  PubMed  Google Scholar 

  29. Garcia-Ovejero D, Veiga S, Garcia-Segura LM, Doncarlos LL. Glial expression of estrogen and androgen receptors after rat brain injury. J Comp Neurol. 2002;450:256–71.

    Article  CAS  PubMed  Google Scholar 

  30. Brown CM, Xu Q, Okhubo N, Vitek MP, Colton CA. Androgen-mediated immune function is altered by the apolipoprotein E gene. Endocrinology. 2007;148:3383–90.

    Article  CAS  PubMed  Google Scholar 

  31. Colton CA, Brown CM, Vitek MP. Sex steroids, APOE genotype and the innate immune system. Neurobiol Aging. 2005;26:363–72.

    Article  CAS  PubMed  Google Scholar 

  32. Raber J. AR, apoE, and cognitive function. Horm Behav. 2008;53:706–15.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  33. Altmann A, Tian L, Henderson VW, Greicius MD. Sex modifies the APOE-related risk of developing Alzheimer disease. Ann Neurol. 2014;75:563–73.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  34. Burwick RM, Ramsay PP, Haines JL, Hauser SL, Oksenberg JR, Pericak-Vance MA, et al. APOE epsilon variation in multiple sclerosis susceptibility and disease severity: some answers. Neurology. 2006;66:1373–83.

    Article  CAS  PubMed  Google Scholar 

  35. Pennacchio LA, Rubin EM. Comparative genomic tools and databases: providing insights into the human genome. J Clin Invest. 2003;111:1099–106.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  36. Constantinescu CS, Farooqi N, O'Brien K, Gran B. Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br J Pharmacol. 2011;164:1079–106.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  37. Sawcer S, Hellenthal G, Pirinen M, Spencer CC, Patsopoulos NA, Moutsianas L, et al. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature. 2011;476:214–9.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  38. Lill CM. Recent advances and future challenges in the genetics of multiple sclerosis. Front neurol. 2014;5:130.

    Article  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to the patients and control individuals participating in this study. We thank Ms. Brit-Maren Schjeide for excellent technical assistance. We thank ICM, Généthon for their help and support. French DNA samples were provided by the BRC-REFGENSEP (BB-0033-00019). We thank K. Hofmann for expert technical assistance.

Disclosure of all authors and co-authors

Study sponsorship or funding: This project was funded by grants from the Cure Alzheimer's Fund (to L.B.) and the German Ministry for Education and Research (BMBF; grant 16SV5538 to L.B., KKNMS to F.Z., grant NBL3 to U.K.Z.). The research leading to these results has received funding from INSERM, AFM and the program “investissements d'avenir” ANR-10-IAIHU-06.

A.S. received personal compensation for activities with Novartis, Sanofi and Almirall Hermal GmbH.

L.A.G has received personal compensations (travel support) from Novartis, Biogen Idec, Genzyme/Sanofi Aventis, Teva, Merck Serono, and Bayer Schering Pharma.

F.Z. has received research grants from Teva, Merck Serono, Novartis and Bayer as well as consultation funds from Teva, Merck Serono, Novartis, Bayer Healthcare, Biogen Idec Germany, ONO, Genzyme and Sanofi Aventis. Her travel compensation has been provided for by the aforementioned companies.

T.K. has received travel expenses and personal compensations from Bayer Healthcare, Teva Pharma, Merck Serono, Novartis, Sanofi Aventis and Biogen Idec as well as grant support from Bayer Schering AG and Novartis.

M.B. received a travel grant from Biogen Idec, travel grants, speaker and consultancy honorarium, and a research grant from Merck Serono GmbH, served on an advisory board and received a travel grant from Almirall Hermal GmbH, and received a travel grant and research grants from Teva GmbH as well as from Novartis Pharma GmbH.

U.Z. received personal compensation and research support from Almirall, Bayer, Biogen Idec, Merck Serono, Novartis, Sanofi Aventis and TEVA.

R.G. has received personal compensation for activities with Bayer Healthcare, Biogen and Teva Neuroscience and in an editorial capacity from Therapeutic Advances in Neurological Disorders, and also received patent payments from Biogen and research support from Bayer Healthcare, Biogen, Merck Serono, Teva Neuroscience, Novartis and from the German Ministry for Education and Research (BMBF, “German Competence Network Multiple Sclerosis” (KKNMS), CONTROL MS, 01GI0914).

A.C. has received personal compensation for activities with Almirall Hermal GmbH, Bayer Schering, Biogen, Merck Serono, Novartis and Teva Neuroscience, research support from Bayer Schering, Biogen, Merck Serono and Novartis and research grants from the German Ministry for Education and Research (BMBF, “German Competence Network Multiple Sclerosis” (KKNMS), CONTROL MS, 01GI0914).

The other authors report no disclosures.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to A. Chan.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

Study concept and design: Chan, Gold, Lill Performance of animal experiments (in vivo, ex vivo): Schrewe, Demir, Böhme Acquisition of human data: Lill, Gerdes, Guillot-Noel, Akkad, Blaschke, Graetz, Hoffjan, Kroner, Rieckmann, Cournu-Rebeix, Zipp, Kümpfel, Buttmann, Zettl, Fontaine, Bertram, Chan Analysis and interpretation of data: Schrewe, Lill, Liu, Bertram, Chan, Salmen, Demir Drafting of manuscript: Schrewe, Lill, Chan, Salmen Critical revision of the manuscript for important intellectual content: Gold, Hermann, Hagemann, ElAli, Gerdes, Guillot-Noel, Akkad, Blaschke, Graetz, Hoffjan, Kroner, Rieckmann, Cournu-Rebeix, Zipp, Kümpfel, Buttmann, Zettl, Fontaine, Bertram Administrative, technical and material support: Chan, Hermann, Hagemann, ElAli, Demir, Böhme All authors read and approved the final manuscript.

Classification: Demyelinating diseases [10]; Animal models [170.020]; Human studies [170.040].

L. Schrewe and C. M. Lill contributed equally to this work.

Additional file

Additional file 1: Figure S1.

Body weight of C57Bl/6 wildtype (apoE +/+) and apoE-deficient (apoE -/-) mice during active MOG35-55 EAE. Figure S2. Histopathological assessment after active MOG35-55 EAE. Table S1. Demographic details of all included patients. Figure S3. Distribution of the MS Severity Score in 3,237 German and French patients. Figure S4. Distribution of the MS Severity Score by rs7412 and rs429358 genotypes in all patients and after sex stratification. (PDF 484 kb)

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schrewe, L., Lill, C., Liu, T. et al. Investigation of sex-specific effects of apolipoprotein E on severity of EAE and MS. J Neuroinflammation 12, 234 (2015). https://doi.org/10.1186/s12974-015-0429-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12974-015-0429-y

Keywords